,V. - . ‘ .3 u w..b.$% if... . fiatmfi y... inevmmwfiw 3.. . . .htMMfi‘. J. f. a»? z 59$ .wmusfirfiz . 1w: . Q” I r” .5?" .7 the I”; u t . .. :Hr A. a hit-JV {L A. 9...“? .. l. . m%w%§fi,fiam§m§.r .; .Vx. 3007 This is to certify that the dissertation entitled THE MECHANOTRANSDUCTION RESPONSE LIBRARY Michigan State University Doctoral OF TENDON CELLS TO TENSILE LOADING presented by MICHAEL LAVAGNINO has been accepted towards fulfillment of the requirements for the degree in Mechanical Engineering aged/AmL ' MEX Professor’s Signature 05:14 9/0 7 Date MSU is an affirmative-action, equal-opportunity employer co-I------o—------I----------v---o------n--.-c-.-----------v---o—----u-o-----—--n-----0-------~--n-n-o---. PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 6f07 plelRC/DateDueindd-p.1 THE MECHANOTRANSDUCTION RESPONSE OF TENDON CELLS TO TEN SILE LOADING By Michael Lavagnino A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Mechanical Engineering 2007 ABSTRACT THE MECHANOTRANSDUCTION RESPONSE OF TEN DON CELLS TO TENSILE LOADING By Michael Lavagnino The ability of tendon cells to sense and respond to load is central to the concept of mechanotransduction and the maintenance of tendon homeostasis. Tendon cells sense load through a mechano—electrochemical sensory system(s) that detects mechanical load signals through the deformation of the cellular membrane and/or the cytoskeleton. This cellular deformation produces tension in the cytoskeleton, which can be sensed by the cell nucleus through a mechano-sensory tensegrity system to elicit a metabolic response. While the precise level (magnitude, frequency/rate, and duration) of mechanobiological stimulation required to maintain normal tendon homeostasis is not currently known, it is very likely that an abnormal level(s) of stimulation may play a role in the etiopathogenesis of tendinopathy. Although tendinopathy has been well described pathologically, the precise etiopathogenesis of this condition remains unsettled. Classically, the etiology of tendinopathy has been linked to the performance of repetitive activities (so-called overuse injuries). This has led many investigators to suggest that it is the mechanobiologic over-stimulation of tendon cells from repetitive loading that is the initial stimulus for the degradative processes that have been shown to accompany tendinopathy. Although several studies have been able to demonstrate that the in vitro over-stimulation of tendon cells in monolayer can result in a pattern(s) of gene expression seen in clinical cases of tendinopathy the strain magnitudes and durations used in these in vitro studies, as well as the model systems, may not be clinically relevant. Using an in vitro rat tail tendon model, the objective of this research was to study the mechanobiologic response of tendon cells in situ (within their normal extracellular matrix), to various tensile loading regimes. The studies have shown that the gene response of tendon cells to load is both frequency and amplitude dependent and that tendon cells appear to be “programmed” to sense a certain level of stress. Model analyses combined with the experimental results have demonstrated that both strain rate and strain amplitude are able to independently alter rat interstitial collagenase gene expression through increases in fluid-flow-induced shear stress and matrix-induced cell deformation respectively. The studies have also shown that the absence of stress has a profound effect on the catabolic response of tendon cells, which in turn decreases the mechanical properties of the tendon independent of its collagen fiber distribution. The studies have shown that isolated fibrillar damage can occur within tendons and produce a localized upregulation of interstitial collagenase in response to altered (decreased) tendon cell stimulation. This weakens the tendon and may put more of the extracellular matrix at risk for further damage with subsequent loading. From these studies the hypothesis is forwarded that the etiopathogenic stimulus for the degenerative cascade that precedes the overt pathologic development of tendinopathy is the catabolic response of tendon cells to mechanobiologic under-stimulation as a result of microscopic damage to the collagen fibers of the tendon. This dissertation is a collection of research involving the response of tendon cells to changing load conditions and the examination of the implications of these responses as a potential etiopathogenic mechanism for the onset of tendinopathy. Copyright by MICHAEL LAVAGNINO 2007 DEDICATION To myjoy, my beloved, my wife, Nicole. ACKNOWLEDGEMENTS I would like to thank God for the many people in my life that have made this dissertation possible. I am especially grateful for: My advisor, Dr. Roger C. Haut, whose guidance and encouragement has helped me through each step of the way. I am thankful for his invaluable expertise that he has shared with me in the field of orthopaedic biomechanics. My mentor, Dr. Steven P. Arnoczky, whose scientific excellence and vision has been the inspiration for my work. I am grateful for his friendship, patience, and generosity in providing the opportunity to pursue this degree. My committee members: Drs. Dahsin Liu and Tom Pence for their time, guidance, and assistance. My co-workers: Keri Gardner, Oscar Caballero, Dr. Monika Egerbacher, Jean Kilfoyle, and Tao Tian for their friendship, talents, and assistance with my research. My fellow students: Katie Frank, Eugene Kepich, Eric Meyer, Shrishail Nashi, Erin Robertson, and Drs. Meghan Burns, Kevin Riutort, Erika Sorge, and Zachary Vaupel for their help. Staff: Ralph Common, Mike McLean, and Shirley Owens for their expertise. Faculty: Drs. Steve Elder, Niell Elvin, Steve Goldstein, Jim Kimura, Robert Malinowski, Steve Shaw, and Joanne Whallon for their consultation, help, and advise. My family-in-law: Michael and Diane Sheehy, D. Johnson, Denise and Jimmy Deardorff, and Steve Sheehy for their prayers and love. vi My extended family: All my uncles, aunts and cousins for their continuous interest and support, especially my Uncle Mike Lavagnino, who always wanted to have a business card with Mike Lavagnino, PhD. My Godparents: Uncle Al and Aunt Betty Lavagnino for their love and example. My brothers and sister-in-law: Steve, Albert, and Ayn Lavagnino who fostered my desire to learn and learn quickly, as well as for their encouragement and example. My parents: John and Anna Lavagnino, for their loving sacrifice, inspiration, and motivation for my education. My children: Clare and John for their joy and laughter. Finally I would like to offer my deepest appreciation to my wife, Nicole, who has been my constant support and encouragement from the beginning. I am grateful for the financial support I received from the Laboratory for Comparative Orthopaedic Research and the Wade 0. Brinker Endowed Chair in the College of Veterinary Medicine at Michigan State University. In addition, I would like to acknowledge the College of Veterinary Medicine at Michigan State University for providing me with a Graduate School Dissertation Completion Fellowship. vii TABLE OF CONTENTS LIST OF TABLES ...................................................... x LIST OF FIGURES .................................................... xi INTRODUCTION ...................................................... 1 CHAPTER 1 — Effect of Amplitude and Frequency of Cyclic Tensile Strain on CHAPTER 2 — CHAPTER 3 — the Inhibition of MMP-l3 mRNA Expression in Tendon Cells: an in vitro study ....................................... 28 ABSTRACT ........................................... 29 INTRODUCTION ...................................... 30 MATERIALS AND METHODS ........................... 31 RESULTS ............................................ 36 DISCUSSION ......................................... 39 REFERENCES ........................................ 44 In vitro Alterations in Cytoskeletal Tensional Homeostasis Control Gene Expression in Tendon Cells ................. 48 ABSTRACT ........................................... 49 INTRODUCTION ...................................... 50 MATERIALS AND METHODS ........................... 52 RESULTS ............................................ 57 DISCUSSION ......................................... 63 ACKNOWLEDGEMENTS ............................... 67 REFERENCES ........................................ 68 Collagen F ibril Diameter Distribution Does Not Reflect Changes in the Mechanical Properties of in vitro Stress-Deprived Tendons ....................... 73 ABSTRACT ........................................... 74 INTRODUCTION ...................................... 75 MATERIALS AND METHODS ........................... 76 RESULTS ............................................ 80 DISCUSSION ......................................... 86 ACKNOWLEDGEMENTS ............................... 90 REFERENCES ........................................ 91 viii CHAPTER 4 — Isolated F ibrillar Damage in Tendons Stimulates Local Collagenase mRNA Expression and Protein Synthesis ....... 95 ABSTRACT ........................................... 96 INTRODUCTION ...................................... 97 MATERIALS AND METHODS ........................... 99 RESULTS ........................................... 104 DISCUSSION ........................................ 107 REFERENCES ....................................... 113 CHAPTER 5 — A Finite Element Model Predicts the Mechanotransduction Response of Tendon Cells to Cyclic Tensile Loading .............................. 116 ABSTRACT ......................................... 117 INTRODUCTION ..................................... 1 18 MODEL DEVELOPMENT .............................. 121 FINlTE ELEMENT METHOD ........................... 124 EXPERIMENTAL METHODS ........................... 130 RESULTS ........................................... 132 DISCUSSION ........................................ 137 REFERENCES ....................................... 144 CONCLUSIONS ..................................................... 150 APPENDIX A — Ex vivo static tensile loading inhibits MMP-l expression in rat tail tendon cells through a cytoskeletally based mechanotransduction mechanism ........................ 153 ABSTRACT .......................................... 154 INTRODUCTION .................................... 155 MATERIALS AND METHODS .......................... 156 RESULTS ........................................... 160 DISCUSSION ........................................ 165 REFERENCES ....................................... 170 APPENDIX B — Computational input files B.1 Global Model for 1% strain at 2% strain/minute .......... 173 8.2 Global Model for 1% strain at 20% strain/minute ......... 180 B3 Global Model for 3% strain at 6% strain/minute .......... 180 B4 Global Model for 3% strain at 2% strain/minute .......... 180 B5 Submodel for 1% strain at 2% strain/minute ............. 181 B6 Submodel for 1% strain at 20% strain/minute ............ 196 B7 Submodel for 3% strain at 6% strain/minute ........... 196 3.8 Submodel for 3% strain at 2% strain/minute ............. 196 ix LIST OF TABLES Table 3.1 Mean : standard deviation for control and stress-deprived fibril number, mean fibril diameter and mean fibril density. Resulting p-value from paired t-test with significance set at p<0.05 ..................... 82 Table 3.2 Comparison of the cross-sectional area, tensile modulus, failure stress, and failure strain of control and stress-deprived rat tail tendons. TCross—sectional area measurements were paired from the same tendon. The control tendon area was used for both groups to calculate failure stress. * significantly different than control specimens, p<0.05 ......... 85 Table 5.1 Global matrix material properties ................................ 127 Table 5.2 Submodel material properties ................................... 130 Table 5.3 Global model and submodel strain and shear stress values ............ 135 Table A.1 The effect of static stress on MMP-l expression .................... 161 LIST OF FIGURES Figure 1 The hierarchical collagen network of tendon (Kastelic et al. 1978) ........ 3 Figure 2 Extension of an elastic collagen fiber zig-zag crimp with apex points of infinite rigidity (Diamant et al. 1972) ............................. 4 Figure 3 Advancements in crimp definition using a blunted zig-zag (b: crimp blunting factor) used in the SSL model (Kastelic et al. 1980). The undeformed and deformed configurations of collagen crimp with spring apex points (Stouffer et al. 1985) ............................. 5 Figure 4 A schematic diagram showing the bilinear stress-strain curve for a single collagen fibril. Fibrils can vary by strain at which the crimp is fully stretched (S) or at which ultimate failure occurs (U) (Kwan and Woo 1989) .......................................... 6 Figure 5 Spring-element model showing nonlinear spring elements (fibers) overlaid on the continuum elements (matrix) (Wilson et a1. 1997) ........ 8 Figure 6 (a) Schematic representation of the hierarchical structure of a collagen tendon. If (a fibre of) the tendon is stretched by an amount 81', this is distributed between the collagen fibrils (cf) with a tensile strain 80 and the proteoglycan-rich matrix (pg), which is mainly sheared. Covalent cross-links between molecules are drawn schematically within the collagen fibrils. (b) An illustration representing a mechanical model, where fibrils and matrix are considered as viscoelastic systems arranged in series. ED is the elastic modulus of the fibrils that depends critically on the covalent cross-links. n0 is the viscosity of the fibrils, possibly due to friction between molecules. EM is the effective elastic modulus of the matrix and 11M is the viscosity of the matrix (Puxkandl et al. 2002). . 9 Figure 7 Conceptualization of the geometry of a fascicle used as the basis of a finite element model (Atkinson et al. 1997) ..................... 11 Figure 8 A finite-element model of tendon as a two-phase linear elastic fiber reinforced composite using the rebar formulation of ABAQUS (Giori et al. 1993) ............................................. 12 Figure 1.1 A: Photograph of the computer-controlled, stepper motor driven, cyclic loading system. The system permits five tendons to be loaded simultaneously while in media. B: Close up view of the testing system showing rat tail tendons in place .................................. 33 xi Figure 1.2 Figure 1.3 Figure 1.4 Figure 2.1 Figure 2.2 Figure 2.3 Figure 2.4 Representative Northern blot gel from the amplitude experiment illustrating the relative expression of MMP—l3 mRNA expression in fresh control tendons (lane 1); immobile for 24 hours (lane 2); 1% cyclic strain at 0.017Hz for 24 hours (lane 3); 3% cyclic strain at 0.017Hz for 24 hours (lane 4); 6% cyclic strain at 0.017Hz for 24 hours (lane 5). GAPDH was used as an internal control. Experiments were performed three times and a representative result is shown ...................... 36 Representative Northern blot gel from the frequency experiment illustrating the relative expression of MMP-l3 mRNA expression in fresh control tendons (lane 1); immobile for 24 hours (lane 2); 1% cyclic strain at 0.017Hz for 24 hours (lane 3); 1% cyclic strain at 0.17Hz for 24 hours (lane 4); 1% cyclic strain at 1.0Hz for 24 hours (lane 5). GAPDH was used as an internal control. Experiments were performed three times and a representative result is shown ...................... 37 Representative Northern blot gel from the cytochalasin D experiment illustrating the relative expression of MMP—13 mRNA expression in fresh control tendons (lane 1); immobile for 24 hours (lane 2); 6% cyclic strain at 0.017Hz for 24 hours (lane 3); 6% cyclic strain at 0.017Hz for 24 hours with 10 uM of cytochalasin D for 24 hours (lane 4). GAPDH was used as an internal control. Experiments were performed three times and a representative result is shown ...................... 38 Photograph showing the 3-point traction devices ..................... 54 Representative rhodamine-phalloidin stained cell images under confocal microscopy (40x) of A) elongated cells in adhered gels at 48 hours containing well-organized actin stress fibers, B) the addition of cytochalasin D to the adhered gels or C) the physical release of the gels from the culture dish resulted in an immediate loss of actin stress fiber organization ............................................. 57 Photograph showing a representative gel after 48 hours of attachment to its culture dish (A) and the contraction of the gel following release after 24 hrs (B), 10 days (C), and 14 days (D). (Scale bar = 10 mm) ......... 58 Representative Northern blot analysis of rat interstitial collagenase and (11(1) collagen with GAPDH as a control. Lanes represent 1) adhered to dish for 24 hours, 2) cytochalasin D for 24 hours, 3) 24 hours contraction, 4) 10 days contraction, 5) 10 days contraction plus cytochalasin D for additional 24 hours, 6) 14 days contraction ...................... 59 xii Figure 2.5 Figure 2.6 Figure 3.1 Figure 3.2 Figure 3.3 Figure 3.4 Figure 3.5 Figure 4.1 Photograph showing a representative gel with a 3—point traction device in place immediately after release (A) and after 24 hrs (B) and 10 days (C) of gel contraction around the three pins. Removal of the traction devices (and the opposing tractional resistance they provided) permitted further contraction of the gels (D). (Scale bar = 10 mm) .............. 61 Representative Northern blot analysis of rat interstitial collagenase and (11(1) collagen with GAPDH as a control. Lanes represent 1) adhered to dish for 24 hours, 2) 24 hours contraction around 3pt traction pins, 3) 10 days contraction around pins, 4) 10 days contraction around pins plus free contraction for an additional 24 hours ...................... 62 Representative transmission electron microscope image of A: control rat tail tendon cross-section and of B: a cross-section of a rat tail tendon stress-deprived for 21 days (x 19,000). Scale bar = 500 nm ........... 81 Histogram illustrating collagen fibril density in control and 21 day stress- deprived rat tail tendons for each of the six paired tendons measured and for all the control and stress-deprived tendons combined. There was no significant differences between tendons, p>0.05 ................... 82 Histogram illustrating relative frequencies of collagen fibril diameters in control and 21 day stress-deprived rat tail tendons. There was no significant differences between tendons within each bin, p>0.05 ........ 83 Representative stress versus strain curves from paired control and 21 day stress-deprived rat tail tendons of one fibril ......................... 84 Northern blot gel illustrating the relative expression of MMP-l3 mRN A expression in fresh control tendons (lane 1) and stress-deprived for 21 days (lane 2). GAPDH was used as an internal control ................ 85 Representative stress-strain curve of a rat tail tendon fascicle (dark solid line) demonstrating the point at which fibrillar damage occurred (point C), and the unloading of the tendon to 100g (dashed line). A representative curve of the tendon loaded to failure displaying the negative slope in the stress strain curve that eventually ends in total tendon failure (Lavagnino et al. 2005) has also been included (light solid line). Points A-D on the stress-strain curve correspond to the images of the fascicle at those points in Figure 4.2A-D ............... 101 xiii Figure 4.2 Figure 4.3 Figure 4.4 Figure 4.5 Figure 5.1 Figure 5.2 Images of a rat tail tendon fascicle at various points throughout the testing protocol: A) Prior to loading (the crimp pattern is clearly visible). B) During loading in the linear portion of the stress-strain curve demonstrating the elimination of the crimp pattern. C) Onset of fibrillar damage as manifested by a change in the reflectivity of the damaged fibrils (arrows). D) Unloading of the tendon to 100g and the reoccurrence of the crimp pattern within the damaged fibrils (arrows). (bar = 200 microns) ............................. 105 Representative images of a rat tail tendon fascicle following fibrillar damage. A) The presence of the crimp pattern on the bottom of the tendon fascicle (arrows) indicates the site of isolated fibrillar damage. B) In situ hybridization of the tendon fascicle reveals interstitial collagenase mRNA expression in those cells associated with the damaged fibril(s). The borders of the tendon fascicle are delineated by broken lines. (bar = 100 microns) ............................. 106 Representative image of a control (unloaded [stress deprived] for 24 hours) rat tail tendon fascicle demonstrating interstitial collagenase mRNA expression by cells throughout the entire fascicle. (bar = lOOmicrons) ........................................... 106 A) Representative photomicrograph of an injured rat tail tendon fascicle showing the damaged fibrils (denoted by the presence of crimp) immediately adjacent to uninjured fibrils. (bar = 20 microns) B) Photornicrograph of the same field under fluorescent light demonstrating the positive (light gray) staining of MMP-13 protein in the cytoplasm of only those cells within the damaged fibrils. The nuclei have been counterstained with DAPI (white) to help identify the cells. (bar = 20 microns) ............................................ 107 Axisymmetric global poroelastic model of the rat tail tendon (20mm x 0.15mm), divided into 300 4-noded axisymmetric elements (0.2mm x 0.05mm) with radially variant nonlinear spring elements attached at the nodes of the matrix elements (springs), zero pore pressure on the outer boundary (circles), constrained at the tendon center (triangles), and loaded at the tendon end as per previous experimental conditions (arrows). The darkened element boundary indicates the location of the submodel ............. 125 Reaction force (N) plotted against strain (%) to show the radial variation in fiber recruitment from the outer boundary to the inner or center that predicts the nonlinear response of the global tendon ...... 126 xiv Figure 5.3 Figure 5.4 Figure 5.5 Figure 5.6 Figure 5.7 Figure 5.8 Figure A.1 Figure A.2 Figure A.3 Figure A.4 Submodel of the rat tail tendon, the size of a global element, composed of an ovoid-shaped cell, cell membrane, pericellular matrix (PCM), extracellular matrix (ECM), and collagen fibers .................... 128 Comparison of tendon model to actual tendon stress-strain response (3% strain at 6% strain/minute) ................................. 133 Plot of the fluid velocity magnitude (mm/s) showing fluid flow in the positive direction (arrow) out of the tendon (3% strain at 6% strain/min). The curved lines (springs) represent the collagen fibers .............. 133 Plots of the fluid velocity resultant around the cell and out of the tendon (3% strain at 6% strain/min) .................................... 134 Graph of shear-stress induced by fluid flow. Note the marked increase in at the polar ends of the cell ................................... 135 Gene expression levels of rat interstitial collagenase (MMP-13) as determined by Real-Time Quantitative PCR. All experimental samples were quantified relative to the fresh (0 hour) control ................. 136 Photograph of the static tensile loading system. Individual tendons were suspended in 50 ml test tubes filled with culture media containing 10% FBS and calibrated stainless steel weights were attached by clips. . . 157 Representative Northern blot gel illustrating the relative expression of MMP-1 mRNA as a function of applied stress for 24 h. G3PDH was used as an internal control. Experiments were performed three times and a representative result is shown .............................. 161 Composite polynomial regression of graph of the three experimental replicates plotting MMP-l mRNA expression (normalized as a ratio of MMP-1 to G3PDH) against static stress. There was a strong (r2 = 0.78) and significant (p < 0.0001) inverse correlation between MMP-l mRNA expression and static stress ..................................... 163 Western blot analysis illustrating the absence of pro-MMP-l and MMP-1 protein expression in freshly harvested rat tail tendons. There was a significant up-regulation of pro-MMP-l and MMP-1 protein expression in the tendon cells after 24 h of in vitro load deprivation .............. 163 XV Figure A.5 Representative Northern blot gel illustrating the relative expression of MMP—1 mRNA in fresh control tendons (lane 1): 24 h load deprived at 0 MPa (lane 2); 24 h at 2.60 MPa static tensile stress (lane 3): 24 h at 2.60 MPa static tensile stress in 10 pM cytochalasin D (lane 4). G3PDH was used as an internal control. Experiments were performed three times and a representative result is shown ..................... 164 Figure A.6 Confocal overlay images of rat tail tendon cells stained with rhodamine phalloidin to label actin filaments. (A) Fresh control tendon: note presence of actin stress fibers (arrows) in cytoskeleton. (B) Tendon treated with 10 uM cytochalasin D for 1 h; note the absence of organized actin stress fibers. (confocal 40x oil immersion: 2x zoom). . . . 164 xvi INTRODUCTION Tendinopathy, a syndrome of tendon pain, localized tenderness, and impaired performance, is a common and major health issue in workers and athletes who perform repetitive activities (Renstrom and Woo 2007). Although the pathology of tendinopathy has been well described, the precise etiopathogenesis of this condition remains unsettled (Renstrom and Woo 2007). Classically, the etiology of tendinopathy has been linked to the performance of repetitive activities (overuse injuries) (Almekinders et al. 1993). A proposed algorithm for the onset of overuse tendinopathy involves altered cell-matrix interactions in response to repetitive loading (Archambault et al. 1995). In this scenario, repeated strains below the injury threshold of the tendon induce degenerative changes in the tendon-matrix composition and organization (Jarvinen et al. 1997; Jones et al. 2006; Jozsa and Kannus 1997). The degeneration of the extracellular matrix leads to a transient weakness of the tissue making it more susceptible to damage from continued loading. This damage then accumulates until the overt pathology of tendinopathy develops (Archambault et a1. 1995). While this is a feasible algorithm for the development of overuse tendinopathy, the precise mechanism(s) which lead to altered cell-matrix interactions have not been described. To better understand the mechanical association between tendon matrix and tendon cells, it is necessary to understand the following: 1) the composition of the tendon extracellular matrix, 2) the contribution(s) of these extracellular components in defining the material properties of the tendon, 3) the mechanisms by which mechanical signals are transmitted through the extracellular matrix to the tendon cells, and 4) the cellular response to these mechanical signals. Tendon composition Tendons are described as soft connective tissues which link bone to muscle and consist of solid (collagen fibers, cells, and matrix constituents of proteoglycans and glycoproteins) and fluid phases (Kannus 2000; Riley 2005; Woo et al. 1997). Type I collagen is the main component of the solid phase (65-80% dry weight) and is organized within the tendon in parallel fiber bundles with hierarchies of fibrillar arrangement down to microfibril size (Kannus 2000; Kastelic et al. 1978) (Figure 1). Under polarized light microscopy, tendon collagen fibrils appear in a sinusoidal wave pattern referred to as crimp (Diamant et al. 1972). Collagen has the ability to form covalent intramolecular and intermolecular cross-links, which are the keys to its tensile strength characteristics and resistance to chemical or enzymatic breakdown (Tanzer 1973; Woo et al. 1997). The matrix, or ground substance that surrounds the collagen, consists of proteoglycans, glycosaminoglycans (GAGs), and glycoproteins. Proteoglycans and GAGs only make up a small percentage of the total dry tendon weight, but due to their highly negative charge, these molecules attract and limit the movement of water, which represents 60-80% of the total wet weight (Woo et al. 1997). This water binding capacity improves the mechanical properties of tendon against shear and compressive forces (Kannus 2000). Glycoproteins in tendons include both structural and adhesive molecules. The structural glycoproteins (elastin, fibrillin) provide elastic properties of tendon while the adhesive glycoproteins mediate cell-matrix interactions (e. g. tenascin-C, fibronectin, thrombospondin)(Riley 2005). Each tendon component (collagen, crosslinks, crimp, matrix, water) has mechanical significance and together they form a well-organized tissue for optimal load distribution and response (Woo et al. 1997). "éii at x; - . 93'. MICRO— SUB- '.-tr.‘;v" TENDON l"'31“! ~ FIBRIL . cm . “3“"? TROPO- keg}? . COLLAGEN -’u’ I 35:153me 640A ‘ penodrcrty reticbular - - mem rane fibroblasts wavg£orm f as ci cul a r I ' I crimp structure membrane I I I 15 A 35 A 100-200 A 500-5000 A 5030011 1005000 ' SIZE SCALE Figure l The hierarchical collagen network of tendon (Kastelic et al. 1978). Tendon Computational Models Based on these structural components (collagen fibers, proteoglycan matrix, and fluid) computational models of tendon have been created to further understand the mechanical response of tendon to applied strain. Although no models have examined the role of tendon mechanical load on cell deformation, many structural and continuum models exist that accurately model overall tendon mechanical behavior. Collagen Fibers Initial structural and microstructural models investigated parameters (orientation, number, distribution) of the collagen fibril, the main solid constituent (70-80% dry weight) of tendon, that relate to tissue morphology to describe the mechanical function of the tendon (Belkoff and Haut 1992; Comninou and Yannas 1976; Diamant et a1. 1972; Kastelic et al. 1980; Kwan and Woo 1989; Lanir 1983; Stouffer et a1. 1985; Viidik 1972). These models assumed that the toe-in region occurs due to a structural feature of the tissue, i.e., the gradual removal of crimp during deformation leads to increasing stiffness. The earliest structural model described tendon as a cable consisting of these crimped strands of microfibrils where the stress behavior is determined by the properties of the individual strands (Diamant et a1. 1972). These microfibrils were modeled as having elastic segments joined by rigid hinges based on the elastica problem in mechanics (Figure 2). A A l (a) A A A Figure 2 Extension of an elastic collagen fiber zig-zag crimp with apex points of infinite rigidity (Diamant et al. 1972). Investigators advanced this model to explain additional structural features of crimp including the disappearance of crimp apices and the variability in crimp angle along the depth and length of the tendon using linearly elastic collagen fibers (Comninou and Yannas 1976; Kastelic et al. 1980; Stouffer et al. 1985) (Figure 3). Figure3 Advancements in crimp definition using a blunted zig-zag (b: crimp blunting factor) used in the SSL model (Kastelic et al. 1980). The undeformed and deformed configurations of collagen crimp with spring apex points (Stouffer et al. 1985). Including crimp angle variations throughout the tendon was first incorporated in the sequential straightening and loading (SSL) model of collagen crimp (Kastelic et al. 1980). This model suggested that crimped fibrils are assumed to have a negligible resistance to extension while loaded fibrils resist deformation through a linear elastic relationship, thus resistance arises only from the elasticity of already straightened fibrils (Kastelic et al. 1980). This distribution of fibril angle in the tendon leads to a sequential fibril straightening and loading that result in the initial nonlinearity of the toe region (Kastelic et al. 1980). Investigators than described crimp using a modified SSL model, with varying initial fiber lengths of a bilinear elastic collagen fiber that have a specified failure stretch/strain (Figure 4) (Belkoff and Haut 1992; Crisco and Panjabi 1996; Hurschler et al. 1997; Kwan and Woo 1989; Liao and Belkoff 1999). A 0' 8: 8s 8u Figure 4 A schematic diagram showing the bilinear stress-strain curve for a single collagen fibril. Fibrils can vary by strain at which the crimp is fully stretched (S) or at which ultimate failure occurs (U) (Kwan and Woo 1989). These models defined varying initial fibril lengths and varying failure lengths'either arbitrarily (Kwan and Woo 1989), based on normal distributions (Crisco and Panjabi 1996), or extensively on the organization of the collagen microstructure (fiber density and distribution) (Hurschler et al. 1997) to determine how collagen affects the material properties. Other studies have further refined the model by using a quasi-linear viscoelastity law incorporating a relaxation function and a nonlinear time independent elastic response for collagen fibers (Decraemer et al. 1980; Frisen et al. 1969; Fung 1967; Haut and Little 1972; Lanir 1980; Viidik and Ekholm 1968). One proposed viscoelastic model, the single integral finite strain (SIFS), utilizes the concept of constitutive branching to allow different constitutive equations at different elongations and the model is fully nonlinear and reduces to classic viscoelasticity (Mooney-Rivlin) if linearized (Johnson et al. 1996). Thus instead of a bilinear elastic model to describe crimp as shown previously (Kwan and Woo 1989), this model describes the change in micromechanism caused by the onset or cessation of collagen fiber recruitment as the tendon is extended as a change in constitutive equations. This model appears very accurate at modeling stress- relaxation, peak cyclic stresses, and stress strain curves (Johnson et al. 1996). Proteoglycan Matrix Although these models accurately depict the whole range of tissue behavior from initial load to failure, by only taking into account collagen structure and properties they lack potential mechanical properties from the remainder of the tendon constituents (extrafibrillar matrix and water). Tendons have been previously modeled as a composite material to incorporate both collagen fiber and matrix properties (Ault and Hoffman 1992; Ault and Hoffman 1992; Luo et a1. 1998; Puxkandl et a1. 2002; Redaelli et al. 2003; Scott 2003; Wilson et al. 1997; Wren and Carter 1998). One such composite model, similar to the pure collagen model proposed by Hurschler (Hurschler et al. 1997), included both extensive fiber and matrix properties (Wren and Carter 1998). This model allowed for uncrimping, stretching, and breaking of collagen fibers, but in addition described the fibers ability to rotate in the matrix as fully, partially, or un- constrained. Another researcher investigated the relationship between the matrix and the fibers and the likelihood of potential “links” between fibers (Wilson et al. 1997). This model utilized a plane stress finite element model with nonlinear spring elements (fibers) overlaid on the continuum elements (matrix) to study the possibility of shear stress within the tissue (Figure 5) (Wilson et al. 1997). The non-linear springs provide the model with the ability to simulate the nonlinear behavior attributed to the crimp of the collagen fibers. 9——\/‘—7——\/‘—0—\/‘——3 o—fifl—o—w—é—w—b Figure 5 Spring-element model showing nonlinear spring elements (fibers) overlaid on the continuum elements (matrix) (Wilson et al. 1997). This model determined that shear transfer may occur from linking between collagen fibers, but unfortunately only small deformations were modeled. The idea that a model should include fiber-fiber and fiber proteoglycan interaction began a subset of models that delve into modeling collagen fibril crosslinks from the molecular standpoint of matrix components, most notably glycosaminoglycan (GAG) chains. A sliding GAG filament model was proposed in which the GAG chain can slide and re-engage and thus provide mechanical stability (Scott 2003). These GAG chains, located on proteoglycans play a role in retaining water as well as creating a bond between collagen fibrils. One model incorporated this fiber-matrix interaction as a composite material with collagen fibrils embedded in a proteoglycan rich matrix, where the matrix is mostly loaded under shear (Figure 6a) (Puxkandl et al. 2002). Deformation in the tendon occurs through extensibility of collagen fibrils and nonfibril (crosslink, matrix) deformation. The composite structure was modeled in series with collagen fibril and proteoglycan matrix as viscoelastic elements taking into account molecular friction, cross links, viscous relaxation, and matrix shearing (Figure 6b) (Puxkandl et al. 2002). Figure 6 (b) collagen fibril pg matrix molecular viscous friction relaxation £0.50 EM.EM n effective elastic modulus and tensile strain due to: W molecular matrix cross-links shearing (a) Schematic representation of the hierarchical structure of a collagen tendon. If (a fibre of) the tendon is stretched by an amount 81, this is distributed between the collagen fibrils (cf) with a tensile strain 8D and the proteoglycan—rich matrix (pg), which is mainly sheared. Covalent cross— links between molecules are drawn schematically within the collagen fibrils. (b) An illustration representing a mechanical model, where fibrils and matrix are considered as viscoelastic systems arranged in series. ED is the elastic modulus of the fibrils that depends critically on the covalent cross-links. 11D is the viscosity of the fibrils, possibly due to friction between molecules. EM is the effective elastic modulus of the matrix and nMis the viscosity of the matrix (Puxkandl et al. 2002). A similar model of proteoglycan links was also proposed with 40% of elongation due to fibril length change, and the remainder suggested due to relative movement of fibrils (Redaelli et al. 2003). Shear stress was assumed responsible for force transfer from fibril to fibril. Fibril length, diameter, and interfibrillar distance were used as variables. Using this model, it was found that a stress-transfer matrix with low elastic modulus was sufficient and an increase in collagen fibril diameter alone cannot explain tendon mechanical properties (Redaelli et a1. 2003). Interstitial fluid flow None of the previously mentioned collagen or collagen-matrix structural models include the permeability of the tendon or look at fluid exudation (Hannafin and Arnoczky 1994) and its role in the stress strain response (Haut and Haut 1997) within the tendon. A general 3D viscoelastic model for fibrous tissue was proposed that takes into account fluid flow, fiber density, and fiber orientation (Lanir 1983). The collagen fibers were assumed to only be subjected to uniaxial strain along the length of the fiber, with no compressive strength and the fluid flow through the matrix was due to a hydrostatic pressure differential (Lanir 1983). The model accurately predicted that the unfolding of the fiber leads to increased hydrostatic pressure or fluid flow through the matrix (Lanir 1983). Multiple continuum theories include tendon permeability and interstitial fluid flow in tendon models (Adeeb et al. 2004; Atkinson et al. 1997; Butler et al. 1997; Chen and Ingber 1999; Yin and Elliott 2004). These continuum models have added the structural morphology of the tendon extracellular matrix with fluid using a biphasic approach (Atkinson et al. 1997; Giori et al. 1993; Luo et al. 1998; Wakabayashi et al. 2003; Yin and Elliott 2004). One such biphasic model assumed that tendon was transversely isotropic in the fiber direction and the fibers are assumed to be piecewise linear elastic (Yin and Elliott 2004). Each of these models has predicted the importance of fluid flow and permeability in determining intertendonous strains and the exudation of 10 fluid flow from the tendon. These theories however do not take into account the nonhomogeneity of the crimp pattern (Hansen et al. 2002) nor the viscoelastic properties of the collagen fibers (Haut and Little 1972). Collagen, matrix, and fluid flow There are relatively few tendon models that take into account collagen structure, and a porous matrix that allows for fluid flow. One finite element model assumed the collagen crimp as an outer helical arrangement of 62° inclination (Figure 7) (Atkinson et al. 1997). The central core was then modeled as nonlinear poroelastic material with varying permeabilities (Atkinson et al. 1997). This model predicted fluid exudation and fiber straightening by having the outer collagen fibers wring out the fluid containing central portion. water based l<737fix106—J matrix sealed w surfaces < / 10.0x10-6 62° collagen / fibers . fiber fixed base orientation Figure 7 Conceptualization of the geometry of a fascicle used as the basis of a finite element model (Atkinson et al. 1997). Another model examining the role of fluid in tissue and potential causes of fluid exudation addressed three mechanisms: 1) crimp straightening, 2) Poisson’s ratio — determined by cross links, and 3) osmotic pressure (Adeeb et al. 2004). Another 11 investigator used a finite element system to track changes in fluid pressure and collagen orientation to examine the alterations in tendon composition with mechanical load (Giori et al. 1993). The maintenance and rearrangement of the tendon’s fibrous extracellular matrix was associated with regions where stretching and distortion of cells take place as the tendon was physiologically loaded (tendon wrapped around bone). The tendon was considered to be a two-phase linear elastic fiber-reinforced composite defined using the rebar formulation of the ABAQUS FEM code (Figure 8) (Giori et al. 1993). This model assumed the tendon had no crimp initially (Giori et al. 1993). Results from this study showed that the hydrostatic stress and distortional strain are important tissue level mechanical stimuli regulating the composition of connective tissue and gene expression (Giori et al. 1993). 16 MPa Average Stress goo-u...— 0.... I "3 0 0‘. Illlllllllllllllllll o IIIIIIIIIIIIIIIIHHP'.‘ llllllllllllllllllll". llllll‘llllllllllllll‘ - mummium - lllllllllllllllllill 9. IIIIIIIIIIIIIIIIIIII (O IIIIIIIIIIIIIIIlIIII . IIIIIIIIIIIIIIIIIIII lIIlIIIIIIIIIIIIlIll IIIIIIIIIIIIIIIIIIII IIIIIIIOIIIIIIIIIIII IIIIIIIIIIIIIIINIII “II"HIIINIIIIIII IIII lllllll IIIIIIIHHIIIIHIIl lllllIl'lIllllllllll III” M" :::: Frictionless :::::: ':': Rigid Surface Figure8 A finite-element model of tendon as a two-phase linear elastic fiber reinforced composite using the rebar formulation of ABAQUS (Giori et al. 1993). 12 Other biphasic tissue studies have suggested the importance of both the fluid and solid phases in the transmission of strain from the tissue to the cell, or mechanotransduction (Baer et al. 2003; Guilak and Mow 2000). Currently there are no tendon models that model the mechanical response of tendon strain using the solid and fluid components of tendon and then take that response to determine what is happening on the cellular level. Although each component is known to play an important role in the mechanical response of tendon to load, their influence on cellular mechanotransduction, or the transmission of strain through the extracellular matrix to the cell, within the tendon is unknown. Cell-Matrix Interactions The ability of tendon cells to sense and respond to a physical stress with a biological response, the concept of mechanotransduction, is vital in maintaining tendon homeostasis (Banes et al. 1995; Ingber 1997; Wang and Ingber 1994). Tendon cells sense physical stress through a mechano-biological sensory system(s) that detects mechanical load signals through the deformation of the cellular membrane and/or the cytoskeleton (Adams 1992; Banes et al. 1995; Brown et al. 1998; Ingber 1997; Wang 2006; Wang et al. 1993; Wang and Ingber 1994; Watson 1991). Cell membrane deformations may open or close stretch-activated ion channels, which control the influx of second-messenger molecules such as calcium and inositol triphosphate (1P3) (Banes et al. 1995; Sachs 1988; Sachs 1988; Shirakura et al. 1995). These second messengers can activate a wide array of cellular machinery including DNA synthesis, mitosis, cell differentiation, and gene expression (Binderman et al. 1984; Ryan 1989; Sachs 1988; Sachs 1988). Cellular deformation also alters the cytoskeletal tension, which in turn, can be sensed by the cell nucleus through a mechano-sensory tensegrity system to elicit a 13 metabolic response (Adams 1992; Arnoczky et al. 2002; Banes et al. 1995; Ben—Ze'ev 1991; Ingber 1997; Wang 2006; Wang et al. 1993; Wang and Ingber 1994; Watson 1991). For both of these signaling pathways, change in cell shape with applied stress is thought to occur through the binding of the cell to extracellular matrix proteins such as collagen and fibronectin (Banes et a1. 1995; Rosales et al. 1995; Sung et al. 1996). The binding between the extracellular matrix proteins and the interior cytoskeleton of the cell occurs through the integrin family of cell surface receptors (Banes et al. 1995; Ingber 1991; Janmey 1998; Rosales et al. 1995; Shyy and Chien 1997; Sung et al. 1996; Wang et al. 1993). Thus mechanotransduction, in response to tendon load, is likely mediated through the deformation of the extracellular matrix, which in turn would result in in situ cell deformation (Banes et al. 1995; Sachs 1988; Watson 1991). The results of a recent study support the hypothesis that mechanical loads placed on tendons result in a concomitant in situ deformation of the cell nucleus (Arnoczky et al. 2002). As has been proposed in cartilage, this nuclear deformation may play a significant role in the mechanotransduction of these tissue loads into intracellular signals (Guilak 1995; Guilak and Mow 2000; Guilak et al. 1995). Mechanotransduction, Homeostasis, and Pathology The importance of load (stress) in the homeostasis of connective tissues has been well documented (Akeson et al. 1974; Hannafin et al. 1995; Noyes 1977; Woo et al. 1982; Woo et al. 1997; Yasuda and Hayashi 1999). Several studies have shown that stress deprivation in ligaments and tendons results in significant alterations in their structural and functional properties (Akeson et al. 1974; Hannafin et al. 1995; Noyes 1977; Woo et al. 1982; Woo et al. 1997; Yasuda and Hayashi 1999). While these 14 alterations in the extra-cellular matrix appear to be cell mediated, the exact mechanism by which tissue load (or lack thereof) affects this process is unclear. Numerous in vitro investigations have demonstrated that when a deformable substrate on which adherent cells have been cultured is cyclically strained this extrinsic deformation activates a wide array of cellular machinery including DNA synthesis, mitosis, gene expression, and cell differentiation (Almekinders et al. 1993; Banes et al. 1994; Banes et al. 1995; Banes et al. 1995; Bhargava et al. 1999; Binderman et al. 1984; Birukov et al. 1995; Brighton et al. 1991; Brown 2000; Cheng et al. 1996; Hsieh et al. 2000; Matyas et al. 1995; Sumpio et a1. 1990). Understanding the relationship between tendon strain and cell deformation may provide insight into the role of physical stress in the homeostasis of normal tissue. While the precise level (magnitude, frequency, and duration) of mechanobiological stimulation required to maintain normal tendon homeostasis in not currently known, it is very likely that an abnormal level(s) of stimulation may play a role in the etiopathogenesis of tendinopathy (Archambault et al. 1995). Over-stimulation Several investigators have suggested that tendinopathy is initiated by repetitive loading which over—stimulates the tendon cells leading to a mechano-biologic response of degeneration (Archambault et al. 2002; Archambault et a1. 2001; Skutek et al. 2001; Tsuzaki et al. 2003; Wang et al. 2003). Over-stimulation of tendon cells in vitro has been shown to induce increases in inflammatory cytokines and degenerative enzymes (Almekinders et al. 1993; Archambault et al. 2002; Banes et al. 1999; Banes et al. 1995; Tsuzaki et al. 2003; Wang et al. 2003). The majority of these in vitro studies are based on the response of large numbers of tendon cells cultured on artificial substrates to various 15 regimes of mechanical loading (Almekinders et al. 1993; Archambault et al. 2002; Arnoczky et al. 2002; Banes et al. 1999; Banes et al. 1995; Tsuzaki et al. 2003). However these monolayer cell cultures may not replicate the normal in situ environmental conditions of tendon cells within a three dimensional, extracellular, collagenous matrix (Arnoczky et al. 2007). Since mechanotransduction signals are known to be mediated through the pericellular matrix to the nucleus via integrin based cell-matrix connections (Banes et al. 1995; Ritty et al. 2003; Sachs 1988; Wang et al. 1993; Watson 1991) it is not clear how, or even if, these complex cell-matrix interactions are maintained or recreated in cell cultures. In addition, the strain magnitudes (> 8%) and durations (> 20 hours) used to elicit an up-regulation in the expression of these inflammatory and catabolic genes may not be clinically relevant (Almekinders et al. 1993; Bhargava et al. 2004; Wang et al. 2003). Because tendon cell strain in situ has been shown to be appreciably less than whole tendon strain (Arnoczky et al. 2002), it is unlikely that such high levels of repetitive tendon cell strain could be reached and maintained in vivo without significant damage occurring within the extracellular matrix of the tendon (Woo 1982). Also, since tendons are known to exhibit non-homogeneous strain patterns in response to tensile load (Kastelic et al. 1978), it would seem impossible to precisely recreate the complex and varied patterns of strain amplitudes experienced by a population of tendon cells in situ by uniformly straining a large population of isolated tenocytes in monolayer. Studies have shown that in rat tail tendons even local tissue strain is nonhomogenous throughout the depth of the tendon (Arnoczky et al. 2002; Hansen et al. 2002). Thus, the in vitro application of high magnitudes of cyclic cellular strain to tendon cells in monolayer for excessively long durations may have little bearing 16 on what is actually occurring to tendon cells in situ and the clinical relevance of these studies must be called into question (Arnoczky et al. 2007). Hypothesis To better understand how the mechanotransduction response of tendon cells under tensile load affects gene expression and may contribute to the etiopathogenesis of tendinopathy, an in Sllll rat tail tendon model has been utilized in an effort to maintain the tendon cells’ natural cell-matrix interactions as well as the naturally occurring strain fields that are developed in response to tensile loading (Arnoczky et al. 2004; Lavagnino et al. 2006; Lavagnino et al. 2005; Lavagnino et al. 2003). In this dissertation the mechanobiological response of tendon cells to changing loading patterns is examined and a hypothesis is forwarded that it is a mechanobiological under-stimulation resulting from altered cell-matrix interactions and not a repetitive over-stimulation of tendon cells that is the etiopathogenic stimulus for the degenerative cascade which may eventually lead to tendinopathy. Chapter Overviews Chapter 1 documents rat interstitial collagenase mRNA expression in an in situ tendon cell model in response to various cyclic loading regimes. In addition, the effect of chemically disrupting a segment of the mechanotransduction mechanism (cytoskeleton) on interstitial collagenase mRNA expression was also examined. This study suggested that removing tendons from their normal mechanical environment could significantly alter the homeostatic tension within the cytoskeletal tensegrity system and be responsible for the up-regulation of collagenase mRN A expression seen following 24 hours of stress- deprivation. In addition, the study demonstrated that interstitial collagenase mRNA 17 expression in tendon cells in situ was inhibited or even eliminated by cyclic tensile strain in a dose-dependent manner (both amplitude and frequency), presumably through a cytoskeletally based mechanotransduction pathway. Chapter 2 investigated the potential that tendon cells may have a threshold, or set- point with regard to their mechanoresponsiveness to tensile loading. A collagen gel matrix model system was used to investigate if changes in the cytoskeletal tensional homeostasis of tendon cells was related to the control of gene expression and to determine the ability of tendon cells to re-establish their cytoskeletal tensional homeostasis in response to a changing mechanical environment. Changes in cytoskeletal tension control a reciprocal expression of anabolic and catabolic genes by tendon cells. Of particular interest in this study was the apparent ability of the tendon cells to re- establish their baseline level of internal cytoskeletal tension (as evidenced by a return to baseline gene expression) following the loss of opposing external forces offered by the collagen matrices following release. Chapter 3 examined if an association exists between the tensile properties and the collagen fibril diameter distribution in in vitro stress-deprived rat tail tendons. The results of this study demonstrated that the decrease in mechanical properties observed in in vitro stress-deprived rat tail tendons was not correlated with the collagen fibril diameter distribution and, therefore, the collagen fibril diameter distribution does not, by itself, dictate the decrease in mechanical properties observed in in vitro stress-deprived rat tail tendons. Chapter 4 examined the ability to create isolated collagen fibril damage and subsequent altered cell-matrix interactions at the damaged site using the rat tail tendon 18 fascicle model. This study demonstrated the creation of isolated tendon fibrillar damage within an otherwise intact tendon fascicle results in an up—regulation of collagenase mRNA expression and protein synthesis by only those tendon cells associated with the damaged fibrils. This would suggest a loss of load-transmitting function in the damaged fibril(s) and a subsequent altered cell-matrix interaction within the affected area. Chapter 5 builds on the experimental study of Chapter 1 with the creation of a multiscale computational tendon model composed of both matrix and fluid phases to examine how global tendon loading may affect fluid-flow-induced shear stresses and membrane strains at the cellular level. The model analysis, combined with additional experimental results, demonstrated that both strain rate and strain amplitude are able to independently alter rat interstitial collagenase gene expression through increases in fluid- flow-induced shear stress and matrix-induced cell deformation respectively. 19 REFERENCES Adams, DS (1992) Mechanisms of cell shape change: the cytomechanics of cellular response to chemical environment and mechanical loading. J Cell Biol 117:83-93. Adeeb, S, Ali, A, Shrive, N, Frank, C and Smith, D (2004) Modelling the behaviour of ligaments: a technical note. Comput Methods Biomech Biomed Engin 7:33-42. Akeson, WH, Woo, SL, Amiel, D and Matthews, JV (1974) Biomechanical and biochemical changes in the periarticular connective tissue during contracture development in the immobilized rabbit knee. Connect Tissue Res 2:315-323. Almekinders, LC, Banes, AJ and Ballenger, CA (1993) Effects of repetitive motion on human fibroblasts. Med Sci Sports Exerc 25:603-607. Archambault, J, Tsuzaki, M, Herzog, W and Banes, AJ (2002) Stretch and interleukin- lbeta induce matrix metalloproteinases in rabbit tendon cells in vitro. J Orthop Res 20:36-39. Archambault, JM, Hart, DA and Herzog, W (2001) Response of rabbit Achilles tendon to chronic repetitive loading. Connect Tissue Res 42: 13-23. Archambault, JM, Wiley, JP and Bray, RC (1995) Exercise loading of tendons and the development of overuse injuries. A review of current literature. Sports Med 20:77-89. Arnoczky, SP, Lavagnino, M and Egerbacher, M (2007) The response of tendon cells to changing loads: Implications in the etiopathogenesis of tendinopathy. In: Tendinopathy in Athletes, SL Woo, P Renstrom and SP Arnoczky (eds.), pp. 46- 59. Oxford, England: Blackwell Publishing. Arnoczky, SP, Lavagnino, M, Whallon, J H and Hoonjan, A (2002) In situ cell nucleus deformation in tendons under tensile load; a morphological analysis using confocal laser microscopy. J Orthop Res 20:29-35. Arnoczky, SP, Tian, T, Lavagnino, M and Gardner, K (2004) Ex vivo static tensile loading inhibits MMP-l expression in rat tail tendon cells through a cytoskeletally based mechanotransduction mechanism. J Orthop Res 22:328-333. Arnoczky, SP, Tian, T, Lavagnino, M, Gardner, K, Schuler, P and Morse, P (2002) Activation of stress-activated protein kinases (SAPK) in tendon cells following cyclic strain: the effects of strain frequency, strain magnitude, and cytosolic calcium. J Orthop Res 20:947-952. Atkinson, TS, Haut, RC and Altiero, NJ (1997) A poroelastic model that predicts some phenomenological responses of ligaments and tendons. J Biomech Eng 119:400- 405. 20 Ault, HK and Hoffman, AH (1992) A composite micromechanical model for connective tissues: Part I--Theory. J Biomech Eng 114:137-141. Ault, HK and Hoffman, AH (1992) A composite micromechanical model for connective tissues: Part II--Application to rat tail tendon and joint capsule. J Biomech Eng 114: 142-146. Baer, AE, Laursen, TA, Guilak, F and Setton, LA (2003) The micromechanical environment of intervertebral disc cells determined by a finite deformation, anisotropic, and biphasic finite element model. J Biomech Eng 12521-11. Banes, AJ, Horesovsky, G, Larson, C, Tsuzaki, M, J udex, S, Archambault, J, Zernicke, R, Herzog, W, Kelley, S and Miller, L (1999) Mechanical load stimulates expression of novel genes in vivo and in vitro in avian flexor tendon cells. Osteoarthritis Cartilage 7: 141-153. Banes, AJ, Sanderson, M, Biotano, S, Hu, P, Brigman, B, Tsuzaki, M, Fischer, T and Lawrence, WT (1994) Mechanical load i growth factors induce [Ca2+]I release, cyclin D1 expression and DNA synthesis in avian tendon cells. In: V Mow, F Guilak, R Tran-Son-Tay and R Hochmuth (Ed), Cell Mechanics and Cellular Engineering. Springer, New York, pp. Pages. Banes, AJ, Tsuzaki, M, Hu, P, Brigman, B, Brown, T, Almekinders, L, Lawrence, WT and Fischer, T (1995) PDGF-BB, IGF-I and mechanical load stimulate DNA synthesis in avian tendon fibroblasts in vitro. J Biomech 28: 1505-1513. Banes, AJ, Tsuzaki, M, Yamamoto, J, Fischer, T, Brigman, B, Brown, T and Miller, L (1995) Mechanoreception at the cellular level: the detection, interpretation, and diversity of responses to mechanical signals. Biochem Cell Biol 73:349-365. Belkoff, SM and Haut, RC (1992) Microstructurally based model analysis of gamma- irradiated tendon allografts. J Orthop Res 10:461-464. Ben-Ze'ev, A (1991) Animal cell shape changes and gene expression. Bioessays 13:207- 212. Bhargava, M, Attia, ET and Hannafin, J A (2004) The effect of cyclic tensile strain on MMPs, collagen, and casein degrading activities of fibroblasts isolated from anterior cruciate and medial collateral ligaments. Transactions of the Orthopaedic Research Society 50:270. Bhargava, MM, Beavis, AJ, Edberg, JC, Warren, RF, Attia, ET and Hannafin, JA (1999) Differential expression of integrin subunits in canine knee ligament fibroblasts. J Orthop Res 17:748-754. Binderman, I, Shimshoni, Z and Somjen, D (1984) Biochemical pathways involved in the translation of physical stimulus into biological message. Calcif Tissue Int 36 Suppl 1:S82-85. 21 Birukov, KG, Shirinsky, VP, Stepanova, OV, Tkachuk, VA, Hahn, AW, Resink, TJ and Smirnov, VN (1995) Stretch affects phenotype and proliferation of vascular smooth muscle cells. Mol Cell Biochem 144: 131-139. Brighton, CT, Strafford, B, Gross, SB, Leatherwood, DF, Williams, JL and Pollack, SR (1991) The proliferative and synthetic response of isolated calvarial bone cells of rats to cyclic biaxial mechanical strain. J Bone Joint Surg Am 73:320-331. Brown, RA, Prajapati, R, McGrouther, DA, Yannas, IV and Eastwood, M (1998) Tensional homeostasis in dermal fibroblasts: mechanical responses to mechanical loading in three-dimensional substrates. J Cell Physiol 175:323-332. Brown, TD (2000) Techniques for mechanical stimulation of cells in vitro: a review. J Biomech 33:3-14. Butler, SL, Kohles, SS, Thielke, RJ, Chen, C and Vanderby, R, Jr. (1997) Interstitial fluid flow in tendons or ligaments: a porous medium finite element simulation. Med Biol Eng Comput 35:742-746. Chen, CS and Ingber, DE (1999) Tensegrity and mechanoregulation: from skeleton to cytoskeleton. Osteoarthritis Cartilage 7:81-94. Cheng, GC, Libby, P, Grodzinsky, AJ and Lee, RT (1996) Induction of DNA synthesis by a single transient mechanical stimulus of human vascular smooth muscle cells. Role of fibroblast growth factor-2. Circulation 93299-105. Comninou, M and Yannas, IV ( 1976) Dependence of stress-strain nonlinearity of connective tissues on the geometry of collagen fibers. J Biomech 92427-433. Crisco, JJ and Panjabi, MM (1996) A Theoretical Model Predicting Ligament Strains. Transactions of the Orthopaedic Research Society 42:52-59. Decraemer, WF, Maes, MA, Vanhuyse, VJ and Vanpeperstraete, P (1980) A non-linear viscoelastic constitutive equation for soft biological tissues, based upon a structural model. J Biomech 13:559-564. Diamant, J, Keller, A, Baer, E, Litt, M and Arridge, RG (1972) Collagen; ultrastructure and its relation to mechanical properties as a function of ageing. Proc R Soc Lond B Biol Sci 180:293-315. Frisen, M, Magi, M, Sonnerup, L and Viidik, A (1969) Rheological analysis of soft collagenous tissue Part 1: Theoretical considerations. Journal of Biomechanics 2: 13-20. Fung, YC (1967) Elasticity of soft tissues in simple elongation. Am J Physiol 213: 1532- 1544. 22 Giori, NJ, Beaupre, GS and Carter, DR (1993) Cellular shape and pressure may mediate mechanical control of tissue composition in tendons. J Orthop Res 11:581-591. Guilak, F (1995) Compression-induced changes in the shape and volume of the chondrocyte nucleus. J Biomech 28:1529-1541. Guilak, F and Mow, VC (2000) The mechanical environment of the chondrocyte: a biphasic finite element model of cell-matrix interactions in articular cartilage. J Biomech 33: 1663-1673. Guilak, F, Ratcliffe, A and Mow, VC (1995) Chondrocyte deformation and local tissue strain in articular cartilage: a confocal microscopy study. J Orthop Res 13:410- 421. Hannafin, J A and Arnoczky, SP (1994) Effect of cyclic and static tensile loading on water content and solute diffusion in canine flexor tendons: an in vitro study. J Orthop Res 12:350-356. Hannafin, JA, Arnoczky, SP, Hoonjan, A and Torzilli, PA (1995) Effect of stress deprivation and cyclic tensile loading on the material and morphologic properties of canine flexor digitorum profundus tendon: an in vitro study. J Orthop Res 13:907-914. Hansen, KA, Weiss, J A and Barton, J K (2002) Recruitment of tendon crimp with applied tensile strain. J Biomech Eng 124272-77. Haut, RC and Little, RW (1972) A constitutive equation for collagen fibers. J Biomech 5:423-430. Haut, TL and Haut, RC (1997) The state of tissue hydration determines the strain-rate- sensitive stiffness of human patellar tendon. J Biomech 30:79-81. Hsieh, AH, Tsai, CM, Ma, Q], Lin, T, Banes, AJ, Villarreal, FJ, Akeson, WH and Sung, KL (2000) Time-dependent increases in type-III collagen gene expression in medical collateral ligament fibroblasts under cyclic strains. J Orthop Res 18:220- 227. Hurschler, C, Loitz-Ramage, B and Vanderby, R, Jr. (1997) A structurally based stress- stretch relationship for tendon and ligament. J Biomech Eng 119:392-399. Ingber, D (1991) Integrins as mechanochemical transducers. Curr Opin Cell Biol 3:841- 848. Ingber, DE (1997) Tensegrity: the architectural basis of cellular mechanotransduction. Annu Rev Physiol 59:575-599. J anmey, PA ( 1998) The cytoskeleton and cell signaling: component localization and mechanical coupling. Physiol Rev 78:763-781. 23 Jarvinen, M, Jozsa, L, Kannus, P, J arvinen, TL, Kvist, M and Leadbetter, W (1997) Histopathological findings in chronic tendon disorders. Scand J Med Sci Sports 7:86-95. Johnson, GA, Livesay, GA, Woo, SL and Rajagopal, KR (1996) A single integral finite strain viscoelastic model of ligaments and tendons. J Biomech Eng 1 18:221-226. Jones, GC, Corps, AN, Pennington, CJ, Clark, IM, Edwards, DR, Bradley, MM, Hazleman, BL and Riley, GP (2006) Expression profiling of metalloproteinases and tissue inhibitors of metalloproteinases in normal and degenerate human achilles tendon. Arthritis Rheum 54:832-842. Jozsa, LG and Kannus, P (1997) Human Tendons: anatomy, physiology, and pathology. In: (Ed.), Human Kinetics, Champaign, IL, pp. Pages. Kannus, P (2000) Structure of the tendon connective tissue. Scand J Med Sci Sports 10:312-320. Kastelic, J, Galeski, A and Baer, E (1978) The multicomposite structure of tendon. Connect Tissue Res 6:1 1-23. Kastelic, J, Palley, I and Baer, E (1980) A structural mechanical model for tendon crimping. J Biomech 13:887-893. Kwan, MK and Woo, SL (1989) A structural model to describe the nonlinear stress-strain behavior for parallel-fibered collagenous tissues. J Biomech Eng 111:361-363. Lanir, Y (1980) A microstructure model for the rheology of mammalian tendon. J Biomech Eng 1022332-339. Lanir, Y (1983) Constitutive equations for fibrous connective tissues. J Biomech 16: 1-12. Lavagnino, M, Arnoczky, SP, Egerbacher, M, Gardner, KL and Burns, ME (2006) Isolated fibrillar damage in tendons stimulates local collagenase mRNA expression and protein synthesis. J Biomech 39:2355-2362. Lavagnino, M, Arnoczky, SP, Frank, K and Tian, T (2005) Collagen fibril diameter distribution does not reflect changes in the mechanical properties of in vitro stress-deprived tendons. J Biomech 38:69-75. Lavagnino, M, Arnoczky, SP, Tian, T and Vaupel, Z (2003) Effect of amplitude and frequency of cyclic tensile strain on the inhibition of MMP—1 mRNA expression in tendon cells: an in vitro study. Connect Tissue Res 44: 181-187. Liao, H and Belkoff, SM (1999) A failure model for ligaments. J Biomech 32: 183-188. Luo, ZP, Hsu, HC, Grabowski, JJ, Morrey, BF and An, KN (1998) Mechanical environment associated with rotator cuff tears. J Shoulder Elbow Surg 7:616-620. 24 Matyas, JR, Anton, MG, Shrive, NG and Frank, CB ( 1995) Stress governs tissue phenotype at the femoral insertion of the rabbit MCL. J Biomech 28:147-157. Noyes, FR (1977) Functional properties of knee ligaments and alterations induced by immobilization: a correlative biomechanical and histological study in primates. Clin Orthop Relat Res 210-242. Puxkandl, R, Zizak, 1, Paris, 0, Keckes, J, Tesch, W, Bernstorff, S, Purslow, P and Fratzl, P (2002) Viscoelastic properties of collagen: synchrotron radiation investigations and structural model. Philos Trans R Soc Lond B Biol Sci 357: 191-197. Redaelli, A, Vesentini, S, Soncini, M, Vena, P, Mantero, S and Montevecchi, FM (2003) Possible role of decorin glycosaminoglycans in fibril to fibril force transfer in relative mature tendons--a computational study from molecular to microstructural level. J Biomech 36: 1555-1569. Renstrom, P and Woo, SL (2007) Tendinopathy: A Major Medical Problem in Sport. In: Tendinopathy in Athletes, SL Woo, P Renstrom and SP Arnoczky (eds), pp. 1-9. Oxford, England: Blackwell Publishing. Riley, G (2005) Chronic tendon pathology: molecular basis and therapeutic implications. Expert Rev Mol Med 7: 1-25. Ritty, TM, Roth, R and Heuser, JE (2003) Tendon cell array isolation reveals a previously unknown fibrillin-2-containing macromolecular assembly. Structure (Camb) 11:1179-1188. Rosales, C, O'Brien, V, Kornberg, L and Juliano, R (1995) Signal transduction by cell adhesion receptors. Biochim Biophys Acta 1242:77-98. Ryan, TJ (1989) Biochemical consequences of mechanical forces generated by distention and distortion. J Am Acad Dermatol 21:115-130. Sachs, F (1988) Ion channels as mechanical transducers. In: Cell Shape: Determinants, Regulation, and Regulatory Role., WD Stein and F Bronner (eds), pp. 63-90. San Diego: Academic Press. Sachs, F (1988) Mechanical transduction in biological systems. Crit Rev Biomed Eng 16:141-169. Scott, JE (2003) Elasticity in extracellular matrix 'shape modules‘ of tendon, cartilage, etc. A sliding proteoglycan-filament model. J Physiol 553:335—343. Shirakura, K, Ciarelli, M, Arnoczky, SP and Whallon, J H (1995) Deformation induced calcium signaling. Transactions of the Combined Orthopaedic Research Societies 2:94. 25 Shyy, JY and Chien, S (1997) Role of integrins in cellular responses to mechanical stress and adhesion. Curr Opin Cell Biol 91707-713. Skutek, M, van Griensven, M, Zeichen, J, Brauer, N and Bosch, U (2001) Cyclic mechanical stretching enhances secretion of Interleukin 6 in human tendon fibroblasts. Knee Surg Sports Traumatol Arthrosc 9:322-326. Stouffer, DC, Butler, DL and Hosny, D (1985) The relationship between crimp pattern and mechanical response of human patellar tendon-bone units. J Biomech Eng 107:158-165. Sumpio, BE, Banes, AJ, Link, GW and Iba, T (1990) Modulation of endothelial cell phenotype by cyclic stretch: inhibition of collagen production. J Surg Res 48:415- 420. Sung, KL, Whittemore, DE, Yang, L, Amiel, D and Akeson, WH (1996) Signal pathways and ligament cell adhesiveness. J Orthop Res 14:729-735. Tanzer, ML (1973) Cross-linking of collagen. Science 180:561-566. Tsuzaki, M, Bynum, D, Almekinders, L, Yang, X, Faber, J and Banes, AJ (2003) ATP modulates load-inducible IL-lbeta, COX 2, and MMP-3 gene expression in human tendon cells. J Cell Biochem 89:556-562. Viidik, A (1972) Interdependence between structure and function in collagenous tissues. In: A Viidik and J Vaust (Ed.), Biology of Collagen. Academic Press, New York, pp. Pages. Viidik, A and Ekholm, R (1968) Light and electron microscopic studies of callagen fibers under strain. Z Anat Entwicklungsgesch 127:154-164. Wakabayashi, I, Itoi, E, Sano, H, Shibuya, Y, Sashi, R, Minagawa, H and Kobayashi, M (2003) Mechanical environment of the supraspinatus tendon: a two-dimensional finite element model analysis. J Shoulder Elbow Surg 12:612-617. Wang, JH (2006) Mechanobiology of tendon. J Biomech 39: 1563-1582. Wang, JH, Jia, F, Yang, G, Yang, S, Campbell, BH, Stone, D and Woo, SL (2003) Cyclic mechanical stretching of human tendon fibroblasts increases the production of prostaglandin E2 and levels of cyclooxygenase expression: a novel in vitro model study. Connect Tissue Res 44:128-133. Wang, N, Butler, JP and Ingber, DE (1993) Mechanotransduction across the cell surface and through the cytoskeleton. Science 260:1124-1 127. Wang, N and Ingber, DE (1994) Control of cytoskeletal mechanics by extracellular matrix, cell shape, and mechanical tension. Biophys J 66:2181-2189. 26 Watson, PA (1991) Function follows form: generation of intracellular signals by cell deformation. Faseb J 5:2013-2019. Wilson, A, Shelton, F, Chaput, C, Frank, C, Butler, D and Shrive, N (1997) The shear behaviour of the rabbit medial collateral ligament. Med Eng Phys 19:652-657. Woo, SL (1982) Mechanical properties of tendons and ligaments. I. Quasi-static and nonlinear viscoelastic properties. Biorheology 19:385-396. Woo, SL, Gomez, MA, Woo, YK and Akeson, WH (1982) Mechanical properties of tendons and ligaments. II. The relationships of immobilization and exercise on tissue remodeling. Biorheology 19:397-408. Woo, SL, Livesay, GA, Runco, TJ and Young, EP (1997) Structure and function of tendons and ligaments. In: Basic Orthopaedic Biomechanics, VC Mow and WC Hayes (eds.), pp. 209-251. Philadelphia: Lippincott-Raven. Wren, TA and Carter, DR (1998) A microstructural model for the tensile constitutive and failure behavior of soft skeletal connective tissues. J Biomech Eng 120:55-61. Yasuda, K and Hayashi, K (1999) Changes in biomechanical properties of tendons and ligaments from joint disuse. Osteoarthritis Cartilage 7: 122-129. Yin, L and Elliott, DM (2004) A biphasic and transversely isotropic mechanical model for tendon: application to mouse tail fascicles in uniaxial tension. J Biomech 37:907-916. 27 CHAPTER 1 Effect of Amplitude and Frequency of Cyclic Tensile Strain on the Inhibition of MMP-l3 mRNA Expression in Tendon Cells: an in vitro study Michael Lavagnino Steven P. Arnoczky Tao Tian Zachary Vaupel From the Laboratory for Comparative Orthopaedic Research College of Veterinary Medicine, Michigan State University, East Lansing, Michigan 48824, USA Lavagnino, M, Arnoczky, SP, Tian, T and Vaupel, Z (2003) Effect of amplitude and frequency of cyclic tensile strain on the inhibition of MMP-1 mRNA expression in tendon cells: an in vitro study. Connect Tissue Res 44:181-187. 28 ABSTRACT To determine the effect of cyclic strain amplitude and frequency on MMP-13 (interstitial collagenase) expression in tendon cells, rat tail tendons (RTT) were immobilized or cyclically displaced to various amplitudes (1, 3, or 6% strain at 0.017 H2) or frequencies (1% strain at 0.017, 0.17, or 1.0 Hz) for 24 hr. Stress-deprivation for 24 hr resulted in a marked upregulation in MMP-l3 expression. Cyclic tensile loading at 0.017 Hz was found to significantly inhibit, but not completely eliminate, MMP—l3 expression at 1% strain. MMP-13 expression was completely eliminated at 3 and 6% strain. Increasing the frequency of application of the 1% strain to 0.17 or 1.0 Hz completely eliminated MMP-13 expression. Disruption of the actin cytoskeleton with cytochalasin D abolished all inhibitory effects of cyclic strain on MMP-13 expression. The results of our study demonstrate that MMP-13 expression in tendon cells can be modulated by varying amplitudes and frequencies of cyclic tensile strain, presumably through a cytoskeletally based mechanotransduction pathway. 29 INTRODUCTION Stress deprivation has been shown to have deleterious effects on the structural and functional prOperties of ligaments and tendons (Amiel et al. 1982; Boorman et al. 1998; Gamble et al. 1984; Goomer et al. 1999; Hannafin et al. 1995; Loitz et al. 1989; Majima et al. 2000; Majima et al. 1994; Noyes 1977). These effects appear to be cell mediated (Hannafin et al. 1995) and are thought to involve interstitial collagenase based alterations of the extracellular matrix (Goomer et al. 1999; Loitz et al. 1989). Previous in vitro studies have implicated a stress-suppressible effect of tensile stress on interstitial collagenase expression in ligaments (Hannafin et al. 1995; Loitz et al. 1989; Majima et al. 2000). A recent study from our lab has demonstrated that in situ stress-deprivation of rat tail tendon cells resulted in an immediate up-regulation of rat interstitial collagenase (MMP-l3) mRNA expression (Arnoczky et al. 2004). Application of static tensile loading produced a dose-dependent inhibition of MMP-l3 mRNA expression through a cytoskeletally based mechanotransduction mechanism (Arnoczky et al. 2004). However, this inhibition was incomplete at the physiological stresses examined (Arnoczky et a1. 2004). In bone, cyclic loading has been shown to be more effective than static loading in inhibiting catabolic activity and stimulating anabolic processes of cells (Burger and Klein-Nulen 1999; Burger and Klein-Nulend 1999; Jacobs et a1. 1998). Cyclic loading of bone produces oscillatory fluid flow within the lacunar/canalicular network (Burger and Klein-Nulend 1999). This fluid flow, and the resulting shear stress, has been shown to be an important physical signal that influences bone cell metabolism and bone adaptations to mechanical loading (Burger and Klein-Nulen 1999; Burger and Klein-Nulend 1999; Hsieh and Turner 2001; Hung et al. 1995; Jacobs et al. 1998; Owan et al. 1997; You et al. 2000). Indeed, in vitro studies have shown that, at low levels (<0.2%) of tissue strain, fluid flow is a more potent stimulator of bone cells than is matrix deformation itself (You et al. 2000). These findings have led to the successful application of low amplitude, high frequency mechanical stimulation in bone to inhibit catabolism (Rubin et al. 2001). Cyclic loading of tendons has been shown to produce interstitial fluid flow (Butler et al. 1997; Chen et al. 1998; Hannafin and Arnoczky 1994; Lanir et al. 1988), which in turn, has been linked to gene expression through a mechanotransduction process involving a cytoskeletal tensegrity system (Archambault et al. 2002). It is possible that lower amplitudes and increased frequency of repetitive cyclic tensile loading may have a more profound effect on maintaining tendon health than higher amplitudes of low frequency or static loading. Therefore, the purpose of this study was to examine the effect of various amplitudes and frequencies of cyclic tensile strain on the regulation of MMP-l3 expression in rat tail tendon cells. It was our hypothesis that cyclic tensile strain will inhibit MMP-13 expression in tendon cells in a dose dependent (amplitude and frequency) manner through a cytoskeletally based mechanotransduction pathway. MATERIALS AND METHODS Drugs and Chemicals Dulbecco’s modified Eagle medium (DMEM), fetal bovine serum (FBS), Ascorbate, gentamicin, and penicillin-streptomycin-fungizone solution were obtained from Gibco (Grand Island, NY, USA). Cytochalasin D, a fungal product that 31 depolymerizes actin filaments of the cytoskeleton, was obtained from SIGMA (St. Louis, MO, USA). Tendon Culture Following institutional animal care and use approval, tendons were obtained from the tails of adult Sprague Dawley rats. The tendons were removed immediately after euthanasia and maintained in DMEM media supplemented with 10% FBS, antibiotic/antimycotic solution, and Ascorbate at 37°C and 10% CO; for the duration of the experiments. Experimental Groups Tendons were divided into groups for three experiments. Each experiment had a zero time control group of fresh rat tail tendons. The first experiment varied cyclic strain amplitude as follows: Group 1: stress-deprived (no strain) for 24 hours; Group 2: 1% cyclic strain at 0.017Hz for 24 hours; Group 3: 3% cyclic strain at 0.017Hz for 24 hours; Group 4: 6% cyclic strain at 0.017Hz for 24 hours. The second experiment varied cyclic strain frequency as follows: Group I: stress—deprived (no strain) for 24 hours; Group 2: 1% cyclic strain at 0.017Hz for 24 hours; Group 3: 1% cyclic strain at 0.17Hz for 24 hours; Group 4: 1% cyclic strain at 1.0Hz for 24 hours. The third experiment evaluated the effect of cytochalasin D (SIGMA) as follows: Group 1: stress-deprived (no strain) for 24 hours; Group 2: 6% cyclic strain at 0.017Hz for 24 hours; and Group 3: 6% cyclic strain at 0.017Hz with lOuM cytochalasin D for 24 hours. There were twenty tendons per group and each experiment was repeated three times. Testing Setup 32 Stress-deprived tendons were kept in a 60mL dish in complete media and conditions as described above for 24 hours. A sawtooth-shaped waveform of cyclic strain was applied to tendons using a custom made, computer-controlled, stepper motor- driven device (Figure 1.1). The grip-to—grip length was set to 40mm using digital calipers Figure 1.1 A: Photograph of the computer-controlled, stepper motor driven, cyclic loading system. The system permits five tendons to be loaded simultaneously while in media. B: Close up view of the testing system showing rat tail tendons in place. 33 The grip-to-grip length was set to 40mm using digital calipers (Mitutoyo, Tokyo, Japan). Tendons were placed in the device until all visible slack was removed to approximate 0% strain. Tendons were then clamped in the grips to prevent slipping before undergoing 1, 3, or 6% cyclic strain with a step size of 25pm and a rate of 0.017, 0.17, or le. At 1% strain, a frequency of 0.017Hz corresponds to a strain rate of 0.033% strain/second and a total of 1440 load events over the 24-hour testing period. At strains of 3 and 6% this strain rate increases to 0.1 and 0.2% strain/second, respectively. Increasing the frequency to 0.17Hz results in a strain rate of 0.33% strain/second and a total of 14,400 load events, whereas a frequency of 1.0Hz corresponds to a strain rate of 2% strain/second and results in a total of 86,400 load events during the 24-hour test period. Tendon Analysis At the end of the experimental period, the unclamped tendon segment (~40 mm) of the cyclically strained tendons and the entire length of the stress-deprived tendons were collected and total cellular RNA was hybridized with a DNA probe for rat interstitial collagenase (MMP-13) generated in our lab and a human GAPDH cDNA control probe. The exposed films were scanned and MMP-l3 expression was quantified by optical density measurements and standardized as a ratio of GAPDH expression. The effect of strain amplitude, strain frequency, and cytochalasin D on MMP-13 expression was evaluated using an ANOVA and Tukey’s post-hoe test. Significance was set at p<0.05. Construction of MMP-13 DNA Plasmid To prepare the probe, MMP-13 DNA (GeneBank M60616) was amplified by polymerase chain reaction (PCR) using rat genomic DNA template. The primers used 34 were 5’-GCC CAT ACA GTT TGA ATA CAG TAT CTG-3’ and 5’-CCA GTT TAA TAA ACA CCA TCT CTT GA-3’. PCR product (1167-bp) was subjected to electrophoresis on 1% agarose gel and recovered using Qiaex H Kit (QIAGEN Inc., Valencia, CA, USA) and then cloned into pCR®2.1-TOPO® plasmid using TOPO TA Cloning kit (Invitrogen Living Science, Carlsbad, CA, USA). The plasmid was transformed into competent E. coli with selection for kanamycin and ampicillin resistance. The construction was confirmed by restriction enzyme digestion (EcoR I, EcoR V, and Hind III) and polymerase chain reaction. RNA Extraction and Northern Blot Analysis Total cellular RNA was isolated from rat tail tendons by the acid guanidine thiocyanate-phenol—chloroform procedure (totally RNA kit, Ambion Inc., Austin, TX, USA). The RNA samples were subjected to electrophoresis on 1.2% agarose gels containing 0.66M formaldehyde and MOPS, then transferred to a nylon membrane (Pierce Corp, Rockford, IL, USA) for 1 hour in TAE at 300mA. Following transfer, the membrane was air—dried and UV cross-linked at 10 Joules/cmz. The MMP-13 probe and human GAPDH cDNA control probe (Clontech Laboratories, Inc., Palo Alto, CA, USA) were labeled with biotin using the North2South Direct HRP Labeling and Detection Kit (Pierce Corp). The RNA blots were hybridized with labeled probes (10ng/ml hybridization solution) at 55°C for 1 hour. The membrane was then washed with 40 ml (~0.5 mL per cmz) 2x SSC/0.1% SDS at 55°C (3 x 5 minutes) and washed with 40 ml (~0.5 mL per cm2) of 2x SSC at room temperature. Following wash, the membrane was incubated with chemiluminescent working solution for 5 minutes and exposed to films for 5-10 minutes. The films were scanned 35 with a laser film digitizer (Lumiscan 75, Lumisys Inc., Sunnyvale, CA, USA) and MMP— 13 mRNA expression was quantitated by optical density measurements using Scion Image (Scion Corporation, Frederick, MD, USA). RESULTS In all experiments, stress-deprivation for 24 hours resulted in a significant (p<0.05) up-regulation of MMP—l3 expression in tendon cells compared to fresh tendons (Figures 1.2-1.4). MMP—13 GAPDH Lane 1 fresh tendon (0 time) Lane 2 no strain -24 hrs Lane 3 1% cyclic strain @0.017Hz -24hrs Lane 4 3% cyclic strain @0.017Hz -24hrs Lane 5 6% cyclic strain @0.017Hz -24hrs Figure 1.2 Representative Northern blot gel from the amplitude experiment illustrating the relative expression of MMP-13 mRNA expression in fresh control tendons (lane 1); immobile for 24 hours (lane 2); 1% cyclic strain at 0.017Hz for 24 hours (lane 3); 3% cyclic strain at 0.017Hz for 24 hours (lane 4); 6% cyclic strain at 0.017Hz for 24 hours (lane 5). GAPDH was used as an internal control. Experiments were performed three times and a representative result is shown. 36 Amplitude A low cyclic strain amplitude of 1% at 0.017Hz resulted in a significant (p<0.05), but incomplete, inhibition of MMP-13 expression. Increasing the cyclic amplitude to 3 or 6% strain at 0.017Hz completely eliminated MMP-13 expression (Figure 1.2). Frequency A low cyclic strain frequency of 0.017Hz at 1% strain again resulted in a significant (p<0.05), but incomplete, inhibition of MMP-l3 expression. Increasing the cyclic frequency to 0.17 or 1.0Hz completely eliminated MMP-13 expression (Figure 1.3). 1 2 3 4 5 MMP—13 - 4. GAPDH O--- - Lane 1 fresh tendon (0 time) Lane 2 no strain -24 hrs Lane 3 1% cyclic strain @0.017Hz -24hrs Lane 4 1% cyclic strain @0.170Hz -24hrs Lane 5 1% cyclic strain @1.000Hz -24hrs Figure 1.3 Representative Northern blot gel from the frequency experiment illustrating the relative expression of MMP-l3 mRNA expression in fresh control tendons (lane 1); immobile for 24 hours (lane 2); 1% cyclic strain at 0.017Hz for 24 hours (lane 3); 1% cyclic strain at 0.17Hz for 24 hours (lane 4); 1% cyclic strain at 1.0Hz for 24 hours (lane 5). GAPDH was used as an internal control. Experiments were performed three times and a representative result is shown. 37 Cytocghalasin D Disruption of the actin cytoskeleton in rat tail tendon cells abrogated the inhibitory effect of cyclic loading on MMP-13 expression. There was no significant (p=0.56) difference in MMP-13 expression between tendons stress—deprived for 24 hours and tendons exposed to 6% cyclic strain at 0.017Hz and lOuM cytochalasin D for 24 hours (Figure 1.4). 1 2 3 4 WIMP-13 - - GAPDH - -.- - Lane 1 fresh tendon (0 time) Lane 2 no strain -24 hrs Lane 3 6% cyclic strain @0.017Hz -24hrs Lane 4 6% cyclic strain @0.017Hz + cytochalasin -24hrs Figure 1.4 Representative Northern blot gel from the cytochalasin D experiment illustrating the relative expression of MMP-13 mRNA expression in fresh control tendons (lane 1); immobile for 24 hours (lane 2); 6% cyclic strain at 0.017Hz for 24 hours (lane 3); 6% cyclic strain at 0.017Hz for 24 hours with 10 uM of cytochalasin D for 24 hours (lane 4). GAPDH was used as an internal control. Experiments were performed three times and a representative result is shown. 38 DISCUSSION Application of load to bone (Brighton et al. 1991; Burger and Klein-Nulen 1999; Hsieh and Turner 2001; Huiskes et al. 2000; Neidlinger—Wilke et al. 1994; Rubin et al. 2001), ligament (Hannafin et al. 2006; Hsieh et al. 2000), and tendon (Arnoczky et al. 2004; Arnoczky et al. 2002; Banes et al. 1995; Hannafin et al. 1995) has been implicated in the maintenance of tissue homeostasis. This is thought to occur through the transfer of tissue strain to the cell cytoskeleton that, in turn, initiates a mechanotransduction signaling response (Banes et al. 1995; Ingber et al. 1995; Sachs 1988). Transmission of tendon strain to the extracellular matrix and cells has not been completely determined, although substrate (extracellular matrix) strain and fluid flow are potential mechanisms (Archambault et al. 2002; Takai et al. 1991). Previous in vitro studies in ligaments and tendons have shown that tensile loading produces substrate (extracellular matrix) strain that, in turn, alters cell shape (Arnoczky et al. 2002; Matyas et al. 1994). Changes in cell shape and the resulting alterations in the actin cytoskeleton are key components in the mechanotransduction response(s) of cells (Banes et al. 1995; Ingber et al. 1995; Sachs 1988; Wang et al. 1993; Watson 1991). A recent study has shown that when static tensile load is applied to rat tail tendons, rat MMP-l (MMP-13) mRNA expression in tendon cells is inhibited in a dose dependent manner (Arnoczky et al. 2004). This amplitude-dependent inhibition of MMP-13 expression appears to correlate with the progressive loss of collagen crimp (from the surface to the center of the rat tail tendon) and the increase in fiber recruitment reported in tendon fascicles with increasing stresses (Hansen et al. 2002). Thus, sequential increases in substrate strain likely result in an increasing number of cells being deformed 39 (Arnoczky et al. 2002). However, because MMP-13 mRNA expression was only inhibited and not totally eliminated with what would appear to be physiologic levels of static stress, substrate deformation may not be the sole factor, or even the most important factor involved in tendon cell signaling and subsequent gene expression with tensile load. Fluid flow and the resultant shear stress are thought to be other important mechanisms for the transmission of tissue strain to cells (You et al. 2000). Shear strain and an up-regulation in gene expression result when musculoskeletal cells in monolayer are exposed to fluid flow (Archambault et al. 2002; Burger and Klein-Nulen 1999; Hung et a1. 1997; Hung et al. 1995; Jacobs et al. 1998; Owan et al. 1997; Xu et al. 2000; You et al. 2000). With bone cells, fluid flow is thought to play a significantly greater role than substrate deformation in activating gene expression (Owan et al. 1997; You et al. 2000). While cyclic strain-induced fluid flow in bone occurs through a patent and well- developed canalicular network, the flow of interstitial fluid in response to cyclic loading is less defined in tendons (Archambault et al. 2002; Hannafin and Arnoczky 1994). Researchers modeling the interstitial fluid flow in tendons have proposed that tensile loading, and the resulting fluid flow, exert a mechanotranduction effect on the tendon cells through shear strain and pressure; however, the exact levels of fluid-induced shear stress have not been identified (Butler et al. 1997; Chen et al. 1998). More research is necessary to determine the precise role of cyclic tensile loading, fluid flow, and shear stress in the mechanotransduction response of tendon cells in situ. In the current study, increasing the cyclic strain frequency totally eliminated MMP-13 mRNA expression at low amplitude strain levels. Similar effects have been reported in other cell types (Yang et al. 1998). Low amplitude (1%) cyclic straining at 40 le is effective in suppressing interstitial collagenase production in human vascular smooth muscle cells (Yang et al. 1998). Whereas the enhanced response of the cells in the current study may be directly related to the increase in frequency of tensile loading, it also may be a result of the increase in strain rate and/or the increase in the total number of load events associated with increases in loading frequency. Frequency, strain rate, and/or number of loading cycles potentially could have a threshold effect on MMP-13 mRNA expression. While the role of strain rate and number of loading cycles on MMP-13 mRNA expression remains to be elucidated, an in vivo study examining the role of cyclic loading in tendon healing suggests increased loading frequency and not an increase in total load events is responsible for improved mechanical properties in healing tissues (Takai et al. 1991). In that study, loading frequencies (0.017 and 0.2Hz) similar to those used in the current study were compared, but the number of loading events for each frequency remained constant (Takai et al. 1991). The authors reported a significant improvement in the mechanical properties of tendons loaded at the higher frequency (Takai et al. 1991). However, the exact mechanism for this improvement (i.e., inhibition of catabolism or stimulation of anabolism) was not determined. The loading frequencies and amplitudes utilized in this study were commensurate with those used in previous in vivo (Majima et al. 2000; Takai et al. 1991) and in vitro (Hannafin et al. 1995; Yang et al. 1998) studies and reflect accepted physiologic levels for normal tendon activity (Viidik 1990). Since tendons are known to exhibit nonhomeogeneous strain patterns in response to tensile load (Kastelic et al. 1978), it is impossible to determine precise amplitudes of strain experienced by the cells based on 41 overall tendon strain. Studies have shown that in rat tail tendons even local tissue strain is nonhomogenous throughout the depth of the tendon (Arnoczky et al. 2002; Hansen et al. 2002). The overall tissue strains used in this study are within the normal functional range of tendons and the response of interstitial collagenase mRNA expression to tensile load reported in the current study is similar to those reported for an in vivo ligament and tendon study (Majima et al. 2000). As stated, the strain-induced mechanotransduction response is thought to be mediated through the cytoskeleton (You et al. 2000). Changes in cell shape, specifically the loss of actin stress fiber organization, has been strongly correlated with collagenase gene expression (Aggeler et al. 1984; Arnoczky et al. 2004). Procollagenase expression in rabbit synovial fibroblasts was induced by treatments that modified cellular actin (Aggeler et al. 1984). Collagenase synthesis was upregulated within six hours of cytoskeletal alteration (Aggeler et al. 1984) and would suggest that a change in the state of actin assembly has a rapid influence on the kinetics of collagenase mRNA expression. In the current study, treatment cytochalasin D, a fungal product that depolymerizes actin filaments, completely abrogated the cyclic strain-induced inhibition of MMP-l3 mRNA expression. These results match similar findings with static load (Arnoczky et al. 2004) and further support the role of a cytoskeletally based mechanosensory tensegrity system in the control of MMP-13 mRNA expression in tendon cells. A limitation of this study is that only MMP-13 mRNA expression and not protein synthesis was examined. Since gene expression may not correlate directly with synthesis of the active enzyme, the association between stress-deprivation induced MMP—13 expression and extracellular matrix degradation was not examined in this study. While 42 stress-deprivation and cyclic load may have an effect on the regulation of other MMPs, previous in vivo studies have suggested that interstitial collagenase appears to be the major matrix metalloproteinase associated with immobilization-induced alterations of tendons and ligaments (Goomer et al. 1999; Majima et al. 2000). Finally, the current study only examined the ability of cyclic load to inhibit the upregulation of the MMP-l3 gene. Future studies are needed to determine the effects of cyclic loading on downregulating MMP-l3 mRNA expression after its expression has been upregulated. The results of our study demonstrate that MMP-13 mRNA expression in tendon cells in situ can be modulated by cyclic tensile strain in a dose-dependent manner (both amplitude and frequency), presumably through a cytoskeletally based mechanotransduction pathway. Understanding the role of exercise on gene expression may help determine optimal exercise protocols for both injured and healthy tissues through the controlled application of load and frequency. It is possible that lower amplitudes and increased frequency of repetitive cyclic tensile loading may have a more profound effect on maintaining tendon health than higher amplitudes of low frequency or static loading. This could lead to advances in overuse injury prevention and optimal rehabilitation protocols following tendon injury and repair. 43 REFERENCES Aggeler, J, Frisch, SM and Werb, Z (1984) Changes in cell shape correlate with collagenase gene expression in rabbit synovial fibroblasts. J Cell Biol 9821662- 1671. Amiel, D, Woo, SL, Harwood, FL and Akeson, WH (1982) The effect of immobilization on collagen turnover in connective tissue: a biochemical-biomechanical correlation. Acta Orthop Scand 53:325-332. Archambault, JM, Elfervig-Wall, MK, Tsuzaki, M, Herzog, W and Banes, AJ (2002) Rabbit tendon cells produce MMP-3 in response to fluid flow without significant calcium transients. J Biomech 35:303-309. Arnoczky, SP, Lavagnino, M, Whallon, J H and Hoonjan, A (2002) In situ cell nucleus deformation in tendons under tensile load; a morphological analysis using confocal laser microscopy. J Orthop Res 20:29-35. Arnoczky, SP, Tian, T, Lavagnino, M and Gardner, K (2004) Ex vivo static tensile loading inhibits MMP-l expression in rat tail tendon cells through a cytoskeletally based mechanotransduction mechanism. J Orthop Res 22:328-333. Arnoczky, SP, Tian, T, Lavagnino, M, Gardner, K, Schuler, P and Morse, P (2002) Activation of stress-activated protein kinases (SAPK) in tendon cells following cyclic strain: the effects of strain frequency, strain magnitude, and cytosolic calcium. J Orthop Res 202947-952. Banes, AJ, Tsuzaki, M, Hu, P, Brigman, B, Brown, T, Almekinders, L, Lawrence, WT and Fischer, T (1995) PDGF-BB, IGF-I and mechanical load stimulate DNA synthesis in avian tendon fibroblasts in vitro. J Biomech 28:1505-1513. Banes, AJ, Tsuzaki, M, Yamamoto, J, Fischer, T, Brigman, B, Brown, T and Miller, L (1995) Mechanoreception at the cellular level: the detection, interpretation, and diversity of responses to mechanical signals. Biochem Cell Biol 73:349-365. Boorman, RS, Shrive, NG and Frank, CB (1998) Immobilization increases the vulnerability of rabbit medial collateral ligament autografts to creep. J Orthop Res 16:682-689. Brighton, CT, Strafford, B, Gross, SB, Leatherwood, DF, Williams, JL and Pollack, SR ( 1991) The proliferative and synthetic response of isolated calvarial bone cells of rats to cyclic biaxial mechanical strain. J Bone Joint Surg Am 732320-331. Burger, EH and Klein-Nulen, J (1999) Responses of bone cells to biomechanical forces in vitro. Adv Dent Res 13:93-98. Burger, EH and Klein-Nulend, J (1999) Mechanotransduction in bone--role of the lacuno-canalicular network. Faseb J 13 Suppl:S 101-1 12. 44 Butler, SL, Kohles, SS, Thielke, RJ, Chen, C and Vanderby, R, Jr. (1997) Interstitial fluid flow in tendons or ligaments: a porous medium finite element simulation. Med Biol Eng Comput 35:742-746. Chen, CT, Malkus, DS and Vanderby, R, Jr. (1998) A fiber matrix model for interstitial fluid flow and permeability in ligaments and tendons. Biorheology 35: 103-1 18. Gamble, JG, Edwards, CC and Max, SR (1984) Enzymatic adaptation in ligaments during immobilization. AmJ Sports Med 12:221-228. Goomer, RS, Basava, D and Maris, T (1999) Effect of stress deprivation on MMP-l gene expression and regulation of MMP-1 promoter in medial collateral and anterior cruciate ligaments (MCL, ACL) and patellar tendon (PT). Transactions of the Orthopaedic Research Society 24:45. Hannafin, JA and Arnoczky, SP (1994) Effect of cyclic and static tensile loading on water content and solute diffusion in canine flexor tendons: an in vitro study. J Orthop Res 12:350-356. Hannafin, J A, Arnoczky, SP, Hoonjan, A and Torzilli, PA (1995) Effect of stress deprivation and cyclic tensile loading on the material and morphologic properties of canine flexor digitorum profundus tendon: an in vitro study. J Orthop Res 13:907-914. Hannafin, J A, Attia, EA, Henshaw, R, Warren, RF and Bhargava, MM (2006) Effect of cyclic strain and plating matrix on cell proliferation and integrin expression by ligament fibroblasts. J Orthop Res 24: 149-158. Hansen, KA, Weiss, J A and Barton, J K (2002) Recruitment of tendon crimp with applied tensile strain. J Biomech Eng 124:72-77. Hsieh, AH, Tsai, CM, Ma, QJ, Lin, T, Banes, AJ, Villarreal, FJ, Akeson, WH and Sung, KL (2000) Time-dependent increases in type-III collagen gene expression in medical collateral ligament fibroblasts under cyclic strains. J Orthop Res 18:220— 227. Hsieh, YF and Turner, CH (2001) Effects of loading frequency on mechanically induced bone formation. J Bone Miner Res 16:918-924. Huiskes, R, Ruimerman, R, van Lenthe, GH and J anssen, JD (2000) Effects of mechanical forces on maintenance and adaptation of form in trabecular bone. Nature 405:704-706. Hung, CT, Allen, FD, Pollack, SR, Attia, ET, Hannafin, J A and Torzilli, PA (1997) Intracellular calcium response of ACL and MCL ligament fibroblasts to fluid- induced shear stress. Cell Signal 9:587-594. 45 Hung, CT, Pollack, SR, Reilly, TM and Brighton, CT (1995) Real-time calcium response of cultured bone cells to fluid flow. Clin Orthop Relat Res 256-269. Ingber, DE, Prusty, D, Sun, Z, Betensky, H and Wang, N (1995) Cell shape, cytoskeletal mechanics, and cell cycle control in angiogenesis. J Biomech 28:1471-1484. Jacobs, CR, Yellowley, CE, Davis, BR, Zhou, Z, Cimbala, JM and Donahue, HJ (1998) Differential effect of steady versus oscillating flow on bone cells. J Biomech 31:969-976. Kastelic, J, Galeski, A and Baer, E (1978) The multicomposite structure of tendon. Connect Tissue Res 6: 1 1-23. Lanir, Y, Salant, EL and Foux, A (1988) Physico-chemical and microstructural changes in collagen fiber bundles following stretch in-vitro. Biorheology 25:591-603. Loitz, BJ, Zernicke, RF, Vailas, AC, Kody, MH and Meals, RA (1989) Effects of short- term immobilization versus continuous passive motion on the biomechanical and biochemical properties of the rabbit tendon. Clin Orthop Relat Res 265-271. Majima, T, Marchuk, LL, Shrive, NG, Frank, CB and Hart, DA (2000) In-vitro cyclic tensile loading of an immobilized and mobilized ligament autograft selectively inhibits mRNA levels for collagenase (MMP-l). J Orthop Sci 5:503-510. Majima, T, Yasuda, K, Yamamoto, N, Kaneda, K and Hayashi, K (1994) Deterioration of mechanical properties of the autograft in controlled stress-shielded augmentation procedures. An experimental study with rabbit patellar tendon. Am J Sports Med 22:821-829. Matyas, J, Edwards, P, Miniaci, A, Shrive, N, Wilson, J, Bray, R and Frank, C (1994) Ligament tension affects nuclear shape in situ: an in vitro study. Connect Tissue Res 31:45-53. Neidlinger-Wilke, C, Wilke, HJ and Claes, L (1994) Cyclic stretching of human osteoblasts affects proliferation and metabolism: a new experimental method and its application. J Orthop Res 12:70-78. Noyes, FR (1977) Functional properties of knee ligaments and alterations induced by immobilization: a correlative biomechanical and histological study in primates. Clin Orthop Relat Res 210-242. Owan, I, Burr, DB, Turner, CH, Qiu, J, Tu, Y, Onyia, JE and Duncan, RL (1997) Mechanotransduction in bone: osteoblasts are more responsive to fluid forces than mechanical strain. Am J Physiol 273:C810-815. Rubin, C, Xu, G and Judex, S (2001) The anabolic activity of bone tissue, suppressed by disuse, is normalized by brief exposure to extremely low-magnitude mechanical stimuli. Faseb J 15:2225-2229. 46 Sachs, F (1988) Mechanical transduction in biological systems. Crit Rev Biomed Eng 16:141-169. Takai, S, Woo, SL, Horibe, S, Tung, DK and Gelberman, RH (1991) The effects of frequency and duration of controlled passive mobilization on tendon healing. J Orthop Res 92705-713. Viidik, A (1990) Structure and function of normal and healing tendon and ligaments. In: Biomechanics of Diarthroidal Joints, VC Mow, A Ratcliffe and SL Woo (eds), pp. 3-38. New York: Springer. Wang, N, Butler, JP and Ingber, DE ( 1993) Mechanotransduction across the cell surface and through the cytoskeleton. Science 260: 1 124-1 127. Watson, PA (1991) Function follows form: generation of intracellular signals by cell deformation. Faseb J 5:2013-2019. Xu, Z, Buckley, M], Evans, CH and Agarwal, S (2000) Cyclic tensile strain acts as an antagonist of IL-1 beta actions in chondrocytes. J Immunol 165:453-460. Yang, JH, Briggs, WH, Libby, P and Lee, RT (1998) Small mechanical strains selectively suppress matrix metalloproteinase-1 expression by human vascular smooth muscle cells. J Biol Chem 273:6550-6555. You, J, Yellowley, CE, Donahue, HJ, Zhang, Y, Chen, Q and Jacobs, CR (2000) Substrate deformation levels associated with routine physical activity are less stimulatory to bone cells relative to loading-induced oscillatory fluid flow. J Biomech Eng 122:387-393. 47 CHAPTER 2 In vitro Alterations in Cytoskeletal Tensional Homeostasis Control Gene Expression in Tendon Cells Michael Lavagnino Steven P. Arnoczky From the Laboratory for Comparative Orthopaedic Research College of Veterinary Medicine, Michigan State University, East Lansing, Michigan 48824, USA Lavagnino, M and Arnoczky, SP (2005) In vitro alterations in cytoskeletal tensional homeostasis control gene expression in tendon cells. J Orthop Res 23: 121 1-1218. 48 ABSTRACT An in vitro collagen gel system was used to determine the effect of alterations in cytoskeletal tensional homeostasis on gene expression in tendon cells. Collagen gel matrices, seeded with rat tail tendon cells, underwent cytochalasin D and gel contraction treatments designed to alter the internal cytoskeletal homeostasis of the cells. Gels were examined for cytoskeletal organization using a rhodamine phalloidin stain for actin. The effect of altered cytoskeletal organization on mRN A expression of a catabolic (interstitial collagenase) and anabolic (alphal(I) collagen) gene was examined using northern blot analysis. Tendon cells in adhered gels demonstrated a highly organized cytoskeleton and showed evidence of alpha1(I) collagen mRNA expression but no evidence of collagenase mRNA expression. Treatment of the attached gel with cytochalasin D disrupted the cytoskeletal organization and resulted in the up-regulation of collagenase mRNA and the inhibition of alpha1(I) collagen mRNA expression. Release of the gels resulted in a cell mediated gel contraction, an immediate loss of cytoskeletal organization, and an mRNA expression pattern similar to that seen with cytochalasin D treatment. Isometric contraction of the gel on itself or around a 3-point traction device resulted in an mRNA expression pattern similar to the adhered gel. Gene expression in the contracted gels could be reversed through chemical cytoskeletal disruption or removal of the traction device which permitted further gel contraction. The results of the study suggest that tendon cells can establish an internal cytoskeletal tension through interactions with their local extracellular environment. Alterations in this tension appear to control the expression of both catabolic and anabolic genes in a reciprocal manner. 49 INTRODUCTION Mechanoresponsiveness is a fundamental feature of all living tissues (Brown et al. 1998; Ingber 1991; Ingber 1997) and tendons are no exception (Banes et al. 1995). Experiments with cultured tendon cells in monolayer confirm that mechanical stresses can regulate a wide variety of cellular processes including signal transduction, gene expression, and proliferation (Almekinders et al. 1993; Archambault et al. 2002; Arnoczky et al. 2002; Banes et al. 1999; Banes et al. 1994; Banes et al. 1995; Tsuzaki et a1. 2003; Tsuzaki et al. 2003). However, the precise level of mechanical load required to initiate (or inhibit) specific cell processes has not been rigorously investigated. Recent in vitro studies have shown that stress deprivation of tendon cells in situ results in an immediate up-regulation of rat interstitial collagenase via a cytoskeletally based mechanotransduction mechanism (Arnoczky et al. 2004; Lavagnino et al. 2003). Conversely, application of a tensile load has been shown to inhibit mRNA expression of interstitial collagenase in a dose dependent manner; presumably, through the same cytoskeletally based mechanism (Arnoczky et al. 2004). These results suggest that tendon cells may have a threshold, or set-point, with regard to their mechanoresponsiveness to tensile loading. Frost first proposed the concept of the mechanostat set-point to explain the mechanoresponsiveness of bone cells in controlling bone mass (Frost 1987). He theorized that bone cells are programmed to sense a certain level of strain induced signals. If the signal was below the set-point the cell would activate catabolic mechanisms that decrease bone mass (Frost 1987). Conversely, if the strain signal exceeded the set-point, anabolic mechanisms would be activated to increase bone mass 50 (Frost 1987). While this concept provides an explanation of how bone mass adapts to gross overloading and underloading, a recent study has suggested that bone cells can also autoregulate their sensitivity to a strain-induced signal by altering their local microenvironment (Rubin et al. 1999). It was theorized that in response to subtle changes in mechanical stress bone cells could actively tune their microenvironment to maintain their idealized strain environment (Rubin et al. 1999). A similar response has been observed in fibroblasts seeded into collagen gels. These cells have been shown to generate a homeostatic contractile force within their extracellular collagenous matrix (Brown et al. 1998). This is achieved through the creation of tension within the internal cytoskeleton via an actomysin filament sliding mechanism (Chen and Ingber 1999; Dickinson et al. 1994; Skalak et al. 1994). The cells reciprocally increased or decreased their endogenous contractions against changes to opposing external loads (Brown et al. 1998). This response allowed the fibroblasts to respond to perceived changes in mechanical loading in a way that maintained tensional homeostasis between the cell and its surrounding extracellular matrix (Brown et al. 1998). It is probable that cytoskeletal tensional homeostasis is the mechanism by which tendon cells establish and attempt to maintain their mechanostat set-point. The purpose of this study was to determine if changes in the cytoskeletal tensional homeostasis of tendon cells are related to the control of gene expression and to determine the ability of tendon cells to re-establish their cytoskeletal tensional homeostasis in response to a changing mechanical environment. Our hypotheses were that tendon cells can generate an internal tensional homeostasis which calibrates the cell with respect to gene expression (rat interstitial collagenase and (11(1) collagen) and that alterations in this 51 internal stress cause a reciprocal change in the expression of catabolic (interstitial collagenase) and anabolic (ul(I) collagen) genes. MATERIALS AND METHODS Cell Culture Rat tail tendon cells were harvested via primary explant cultures from adult Sprague-Dawley rats euthanized for another unrelated study. The cells were expanded to passage 3 in 75 cm2 tissue culture flasks in Dulbecco’s modified Eagle medium, 10% fetal bovine serum, Ascorbate (150 mg/ml), 0.01 mg/ml gentamicin, and 1% antibiotic/antimycotic solution (Gibco, Grand Island, NY, USA) at 37°C in a 10% C02 atmosphere. Collagen Gel Collagen gels made of 2.4 mg/ml type 1 bovine collagen (Vitrogen, Cohesion Technologies, Palo Alto, CA, USA) were seeded with rat tail tendon cells at a concentration of 400,000 cells/mL. Two milliliter gels were created in individual 60 mm culture dishes and allowed to adhere to the culture dishes for at least 24 hours before treatment. The gels were incubated in complete media composed of Dulbecco’s modified Eagle medium, 10% fetal bovine serum, Ascorbate, and penicillin-streptomycin- fungizone (Gibco, Grand Island, NY, USA) at 37°C and 10% C03. The gels were divided into groups to examine the effects of either chemical or physical treatments on cytoskeletal organization and subsequent gene expression. At the end of the study, gels were frozen at —80°C until processed for Northern blot analysis. Each group consisted of five gels and was repeated three times. Additional gels were used to examine cytoskeletal 52 organization in attached, cytochalasin D treated, and released gels using a rhodamine phalloidin stain. Contraction and Chemical Alteration of the Cytoskeletal Tension Seeded collagen gels were allowed to attach to the culture dishes for 24 hours and then treated as follows: Group 1: left attached to the culture dishes for an additional 24 hours; Group 2: attached to the culture dishes for an additional 24 hours while exposed to 10 uM cytochalasin D (SIGMA, St. Louis, MO, USA) to disrupt the cellular cytoskeleton; Group 3: released from the culture dishes and allowed to contract for 24 hours; Group 4: released and allowed to contract for 10 days; Group 5: released from the culture dishes and allowed to contract for 10 days, and then exposed to 10uM cytochalasin D to disrupt the cellular cytoskeleton for an additional 24 hours; Group 6: released from the culture dishes and allowed to contract for 14 days. Contraction and Physical Alteration of the C ytoskeletal Tension To study response of tendon cells to changing extracellular environments an external 3-point traction device was inserted into the gels immediately following release of the gels from the bottom of the culture dishes, such that the gels contracted around the three stainless steel pins (Figure 2.1). The pins in the device were arranged in an equilateral triangular fashion with 12 mm between each pin. This experiment consisted of allowing seeded collagen gels to attach to the culture dishes for 24 hours and then treating them as follows: Group 1: left attached to the culture dishes for an additional 24 hours; Group 2: released from the culture dishes and allowed to contract for 24 hours around the 3-point traction pins; Group 3: released from the culture dishes and allowed to contract for 10 days around 3-point traction pins; Group 4: released from the culture 53 dishes, allowed to contract for 10 days around 3-point traction pins, and then removal of the pins from the gel allowing for an additional 24 hours of contraction. Figure 2.1 Photograph showing the 3-point traction devices. Construction of DNA plasmids to rat interstitial collagenase and rat at] (I) collagen To prepare the collagenase probe, rat interstitial collagenase DNA (GeneBank M60616) was amplified by polymerase chain reaction using rat genomic DNA template. The primers used were 5’-GCC CAT ACA GTT TGA ATA CAG TAT CTG-3’ and 5’- CCA G'I'I‘ TAA TAA ACA CCA TCT CT'I‘ GA-3’. PCR product (1167-bp) was subjected to electrophoresis on 1% agarose gel and recovered using Qiaex II Kit (QIAGEN Inc., Valencia, CA, USA) and then cloned into pCR®2.l-TOP0® plasmid using TOPO TA Cloning kit (Invitrogen Living Science, Carlsbad, CA, USA). The plasmid was transformed into competent E. coli with selection for kanamycin and ampicillin resistance. The construction was confirmed by restriction enzyme digestion (EcoR I, EcoR V, and Hind HI) and polymerase chain reaction. 54 To prepare the collagen probe, rat (11(1) collagen DNA (GeneBank M60616) was amplified by reverse transcription-polymerase chain reaction with total RNA purified from cultured rat tail tendon fibroblasts. The oligonucleotide primers used were 5’-GTC CAT TCC GAA TTC CTG GTC—3’ and 5’GTC GCA CTG GCG ATA GTG G-3’. PCR product (866-bp) was subjected to electrophoresis on 1% agarose gel and recovered using a Qiaex II Kit (QIAGEN Inc., Valencia, CA, USA), and then cloned into pZErOTM-2 plasmid using a Zero BackgroundTM/Kan Cloning kit (Invitrogen Living Science, Carlsbad, CA, USA). The plasmid was transformed into competent E. coli with selection for kanamycin resistance. The construction was confirmed by restriction enzyme digestion (ECOR I and Hind III) and polymerase chain reaction. RNA Extraction and Northern Blot Analysis A Northern blot analysis was performed to assay for rat interstitial collagenase mRNA expression and (11(1) collagen mRNA expression. The gels within each experimental group were combined and total cellular RNA isolated by the acid guanidine thiocyanate-phenol-chloroform procedure (totally RNA kit, Ambion Inc., Austin, TX, USA). The RNA samples were subjected to electrophoresis on 1.2% agarose gels containing 0.66M formaldehyde and MOPS, then transferred to a nylon membrane (Pierce Corp, Rockford, IL, USA) for 1 hour in TAE at 300 mA. Following transfer, the membrane was air—dried and UV cross-linked at 10 J/cmz. The rat interstitial collagenase probe and (11(1) collagen probe, as well as a human GAPDH cDNA control probe (Clontech Laboratories, Inc., Palo Alto, CA, USA) were labeled with biotin using the North2South Direct HRP Labeling and Detection Kit (Pierce Corp, Rockford, IL, USA). The RNA blots were hybridized with labeled probes (10 55 ng/ml hybridization solution) at 55°C for 1 hour. The membrane was then washed with 40 mL (~ 0.5 mL/cmz) 2x SSC/0.1% SDS at 55°C (3x 5 minutes) and washed with 40 mL (~ 0.5 mL/cmz) of 2x SSC at room temperature. Following the wash procedure, the membrane was incubated with chemiluminescent working solution for 5 minutes and exposed to films for 5-10 minutes. C ytoskeletal Organization To evaluate alterations in cytoskeletal organization following chemical or physical manipulations, additional gels were prepared for actin staining and confocal laser microscopy. Gels were fixed in 10% phosphate buffered formalin and stained with rhodamine phalloidin (5 units/mL) (Molecular Probes, Eugene, Oregon, USA) to examine the actin filament structure of the cells after the following treatments: a gel adhered to a culture dish for 48 hours, a gel adhered to a culture dish for 24 hours and then exposed to lOum cytochalasin D for 1 hour, and a gel adhered to a culture dish for 24 hours and then released and allowed to contract for 5 minutes. The gels were wet mounted on a glass slide for viewing with a Zeiss Pascal Laser Scanning Confocal microscope (Carl Zeiss, Thornwood, NY, USA). Observations were made using a 40x oil immersion objective without a coverslip. Fluorescent images were obtained using a HeNe 543 nm laser with a long pass 560 nm emission filter. Cell shape and actin stress fiber organization were observed in cells throughout the gel. After localizing a representative cell in each gel, a 2x zoom was used to obtain an image. 56 RESULTS Cytoskeletal Organization In gels adhered to the culture dish for 48 hours, the tendon cells appeared elongated and their cytoskeletons contained well-organized actin stress fibers (Figure 2.2A). The addition of cytochalasin D to the adhered gels or the physical release of the gels from the culture dish resulted in an immediate loss of this actin stress fiber organization (Figure 2.2B and C). Figure 2.2 Representative rhodamine-phalloidin stained cell images under confocal microscopy (40x) of A) elongated cells in adhered gels at 48 hours containing well-organized actin stress fibers, B) the addition of cytochalasin D to the adhered gels or C) the physical release of the gels from the culture dish resulted in an immediate loss of actin stress fiber organization. 57 Contraction and Chemical Alteration of the Cytoskeletal Tension Upon release from their attachment to their individual culture dishes, the tendon cell seeded collagen gels were contracted by the tendon cells (Figure 2.3). The contraction continued over the next two weeks condensing the gel into a dense, circular bead at 14 days (Figure 2.3). Figure 2.3 Photograph showing a representative gel after 48 hours of attachment to its culture dish (A) and the contraction of the gel following release after 24 hrs (B), 10 days (C), and 14 days (D). (Scale bar = 10 mm). In each of the groups examined, it was apparent that there was a reciprocal relationship between rat interstitial collagenase and (11(1) collagen mRNA expression such that when one was expressed, the other was not (Figure 2.4). Tendon cells in adhered gels demonstrated expression of (11(1) collagen mRNA but no measurable expression of rat interstitial collagenase mRNA (Lane 1). Disruption of cytoskeletal stress fiber organization from exposure to cytochalasin D (lane 2) or gel release and 58 contraction for 24 hours (lane 3) produced an up-regulation of rat interstitial collagenase mRNA expression and an inhibition of (11(1) collagen mRNA expression compared to the adheredgel. D i: '7: 8 m D 0 (u (u (n 0 (n 'o E 2 8 E. 8 09 .c a: — O — d) O h L. + L. 5 s s s s a < 0 N 1- 1- 1- ' m Interstitial Collagenase mRNA C .3 .CC C GAPDH Type I a1 - W i Collagen mRNA ...CCCGAPDH 1 2 3 4 5 6 Figure 2.4 Representative Northern blot analysis of rat interstitial collagenase and (11(1) collagen with GAPDH as a control. Lanes represent 1) adhered to dish for 24 hours, 2) cytochalasin D for 24 hours, 3) 24 hours contraction, 4) 10 days contraction, 5) 10 days contraction plus cytochalasin D for additional 24 hours, 6) 14 days contraction. 59 Following release and 10 days of contraction (lane 4) the gels became more condensed and the cells demonstrated a down-regulation of interstitial collagenase mRNA expression and an up-regulation of (11(1) collagen mRNA expression (compared to actively contracting gels (lane 3)). The disruption of the cytoskeleton with cytochalasin D following 10 days of gel contraction (lane 5) produced the same mRNA expression pattern seen following cytoskeletal alteration in adhered gels (lane 2). This demonstrated that the cells still possessed the ability to respond to cytoskeletal alteration after 10 days. In gels that were released and allowed to contract for 14 days (lane 6), the gels reached an asymptotic contraction (based on previous studies (Arnoczky et al. 2004)) and the mRNA expression of rat interstitial collagenase and (11(1) collagen was similar to the adhered gel. Contraction and Physical Alteration of the C ytoskeletal Tension As noted previously, the tendon seeded collagen gels began to contract upon release from their individual culture dishes. However, instead of contracting into dense beads the gels contracted around the 3-point traction devices forming equilateral triangular structures comprised of bands of condensing collagen gel (Figure 2.5). Removal of the traction devices (and the opposing tractional resistance they provided) permitted further contraction of the gels (Figure 2.5). 60 Figure 2.5 Photograph showing a representative gel with a 3-point traction device in place immediately after release (A) and after 24 hrs (B) and 10 days (C) of gel contraction around the three pins. Removal of the traction devices (and the opposing tractional resistance they provided) permitted further contraction of the gels (D). (Scale bar = 10 mm). In each of the groups examined, it was apparent that there was a reciprocal relationship between the rat interstitial collagenase mRNA and (11(1) collagen mRNA (Figure 2.6). As noted above, gels adhered to the culture dish for 24 hours (lane 1) produced no measurable expression of rat interstitial collagenase mRNA and a positive expression of (11(1) collagen mRNA. Releasing the gel and allowing it to contract around the 3-point pins for 24 hours (lane 2) caused an immediate up-regulation of rat interstitial collagenase and an inhibition of (11(1) collagen mRNA expression compared to the adhered gel. After releasing the gel and allowing a steady state of contraction around the 3—point pins after 10 days (lane 3) the cells regained similar mRNA expression to the adhered gel. The removal of the 3-point pins after 10 days of gel contraction (lane 4) 61 allowed additional contraction to occur (Figure 2.5). This apparent alteration in cell matrix interaction resulted in an up-regulation in the expression of interstitial collagenase and an inhibition of (11(1) collagen mRNA expression. (D 2 m c ._ 0’ '5. U a IE '5 o 09 h B 3 < N 1- 1- + V}, D Interstitial Collagenase mRNA GAPDH Type I (11 A Collagen mRNA . GAPDH 1 2 3 4 Figure 2.6 Representative Northern blot analysis of rat interstitial collagenase and (11(1) collagen with GAPDH as a control. Lanes represent 1) adhered to dish for 24 hours, 2) 24 hours contraction around 3pt traction pins, 3) 10 days contraction around pins, 4) 10 days contraction around pins plus free contraction for an additional 24 hours. 62 DISCUSSION The results of the current study suggest that changes in cytoskeletal tension control a reciprocal expression of anabolic and catabolic genes by tendon cells. It has been suggested that the cellular regulation of biological function lies in the ability of cells to sense, generate, and balance mechanical forces (Chicurel et al. 1998). This mechanoresponsiveness has been shown to be mediated through a tensegrity apparatus comprised of the cell’s cytoskeleton as well as its attachment(s) to the extracellular matrix (Chen and Ingber 1999; Ingber 1991; Ingber 1997; Kolodney and Wysolmerski 1992; Pourati et al. 1998; Ralphs et al. 2002; Roy et al. 1999; Tomasek et al. 1992). Previous studies have shown that cells can generate an internal tension within their cytoskeleton by way of an actomysin filament sliding mechanism (Dickinson et a1. 1994; Skalak et al. 1994). This mechanism allows a cell to maintain a constant cytoskeletal tension in response to changes in external loading (Brown et al. 1998; Sims et al. 1992; Takakuda and Miyairi 1996). This has been termed cytoskeletal homeostasis and is thought to be the mechanism by which a cell maintains a pre-set level of sensitivity to external loading (Brown et al. 1998). In the current study, a previously described collagen gel matrix model system was used to examine the effect of cytoskeletal tension on gene expression (Brown et al. 1998; Brown et al. 1996; Eastwood et al. 1994; Eastwood et al. 1998; Eastwood et al. 1996; Fringer and Grinnell 2001; Grinnell 1994; Grinnell 1999; Grinnell et al. 1999; He and Grinnell 1994; Lee et al. 1993; Lin and Grinnell 1993; Rosenfeldt et al. 1998). In this system, tendon cells seeded into the collagen gels were able to establish a cytoskeletal tensional homeostasis through an isometric contraction against collagen gel matrices left 63 attached to their culture dishes. This was characterized by the presence of organized stress fibers within the cytoskeleton and an up-regulation of the anabolic gene (0t1(I) collagen). Loss of cytoskeletal organization through chemical disruption or a detachment of the gel resulted in an up-regulation in the expression of the catabolic gene (interstitial collagenase) and an inhibition in the expression of the anabolic gene (0tl(I) collagen). Previous studies have shown that alterations in cell shape, secondary to cell detachment (Aggeler et al. 1984; Unemori and Werb 1986), or loss of extracellular matrix tension produce an increase in interstitial collagenase expression (Arnoczky et al. 2004; Goomer et al. 1999; Lambert et al. 1992; Langholz et a1. 1995; Lavagnino et al. 2005; Lavagnino et al. 2003; Loitz et al. 1989; Mauch et al. 1989; Prajapati et al. 2000; Unemori and Werb 1986) and a decrease in collagen production (Lambert et al. 1992; Mauch et al. 1989; Mochitate et a1. 1991; Prajapati et al. 2000; Unemori and Werb 1986). Detachment of the tendon cell seeded collagen gels was followed by a contraction of the matrices by the cells, either on themselves or around the three point traction devices. The phenomenon of collagen gel matrix contraction by fibroblasts has been documented in numerous studies (Brown et al. 1998; Brown et al. 1996; Eastwood et a]. 1994; Eastwood et a1. 1998; Eastwood et a1. 1996; Fringer and Grinnell 2001; Grinnell 1994; Grinnell 1999; Grinnell et al. 1999; He and Grinnell 1994). The ability of the cells to re-align their extracellular matrix by way of an internal contractile mechanism has been implicated in the process of wound contracture and connective tissue morphogenesis (Grinnell 1994; Grinnell 1999; Grinnell 2000; Harris et al. 1981; Stopak and Harris 1982). In both instances, interstitial collagenase is thought to play a critical role in the remodeling process of the extracellular matrix (Stemlicht and Werb 2001; 64 Werb and Chin 1998). During contraction of the gels the cells are thought to exert an isotonic force against the contracting extra-cellular matrix (Grinnell 2000). As contraction slows and the remodeling matrix is able to produce an opposing external force (either by contracting against fixed points or condensing upon itself) the forces exerted by the cells becomes isometric and a homeostatic internal tension develops (Brown et a1. 1998; Grinnell 2000). In the current study, interstitial collagenase expression was partially inhibited after 10 days in the freely contracting collagen matrices as the gels condensed and began to provide resistance to the cellular tension and completely inhibited after 14 days when the freely contracting gels reached an asymptotic level of contraction (Arnoczky et al. 2004). Contraction of the gels around the 3-point traction devices at 10 days resulted in a complete inhibition of interstitial collagenase. Removal of the traction devices permitted additional gel contraction and initiated an immediate up-regulation of interstitial collagenase expression. In all cases, a decrease in interstitial collagenase expression was accompanied by a reciprocal increase in 0t1(I) collagen expression. Thus, the cells were able to return to their baseline level of gene expression (seen in the attached collagen gel matrices) by altering their local extracellular environment through contraction of the gels. The ability of cells to actively “tune” their microenvironment in order to manipulate the degree to which they sense strain is thought to be the mechanism by which tissues adapt to large variations in mechanical loading over time (Frost 1987; Rubin et al. 1999). A recent study in bone suggested that osteocytes can actively modulate their response to external loading by regulating their connections to the extracellular matrix in both a positive (collagen) and negative (collagenase) manner 65 (Rubin et a1. 1999). It was theorized that through this “autoregulation” the osteocytes were able to adjust their exposure to strain stimulus through the regulation of the physical constraints of their own microenvironment (Rubin et al. 1999). This would permit a cell to maintain a “set-point” of activation with respect to the level of mechanical stress required to initiate a biologic response. This has been termed the “mechanostat set-point” in bone cells (Frost 1987) and a similar phenomenon may occur in tendon cells. Of particular interest in this study was the apparent ability of the tendon cells to re—establish their base-line level of internal cytoskeletal tension (as evidenced by a return to baseline gene expressions) following the loss of opposing external forces offered by the collagen matrices following release. This suggests that tendon cells can play an active role in “recalibrating” their sensitivity to changes in external stresses. While this “recalibration” was accomplished by a gross reorganization and contraction of the pliable collagen gel matrices over a 10 day time period such alterations in cell-matrix interactions may be more localized (i.e. the pericellular matrix) and/or require more time (chronic exposure to altered matrix strains) in a mature connective tissue setting. The current study only examined the response of tendon cells to alterations in cytoskeletal tensional homeostasis caused by a decrease in opposing external forces. The application of increased external forces (above the presumed set-point of the tendon cells) was not examined. A previous study has suggested that dermal fibroblasts can respond to increases in opposing external matrix strains by a reciprocal decrease in contractile force, which maintains a homeostatic tension (Brown et al. 1998). The same may hold true for tendon cells. However, the upper and lower limits of the external forces against which the cell can maintain tensional homeostasis is likely dependent on a myriad of factors 66 including cell type and local extracellular matrix composition as well as the frequency and rate of external stress application. The relationship between internal cytoskeletal tension and gene expression would appear to be a key factor in understanding the ability (or inability) of cells to adapt their extracellular matrix to changing external stresses. The results of this study suggest that tendon cells can establish an internal cytoskeletal tension through interactions with their local extracellular environment. Alterations in this tension appear to control the expression of both catabolic and anabolic genes in a reciprocal manner. Although mechanotranduction signals are mediated through a variety of means (focal adhesions, integrins, cytoskeleton) (Banes et al. 1995), the current study focused on how cytoskeletal transformations lead to changes in gene expression. Additional studies are needed to examine the role of cell-matrix connections in establishing and maintaining tensional homeostasis in tendon cells and to determine the precise mechanical/physical threshold (set-point) that is required to activate various catabolic and anabolic genes. ACKNOWLEDGEMENTS The authors would like to thank Keri Gardner and Tao Tian, PhD for their technical expertise. 67 REFERENCES Aggeler, J, Frisch, SM and Werb, Z (1984) Changes in cell shape correlate with collagenase gene expression in rabbit synovial fibroblasts. J Cell Biol 98: 1662- 1671. Almekinders, LC, Banes, AJ and Ballenger, CA (1993) Effects of repetitive motion on human fibroblasts. Med Sci Sports Exerc 25:603—607. Archambault, J, Tsuzaki, M, Herzog, W and Banes, AJ (2002) Stretch and interleukin- lbeta induce matrix metalloproteinases in rabbit tendon cells in vitro. J Orthop Res 20:36-39. Arnoczky, SP, Lavagnino, M, Gardner, KL, Tian, T, Vaupel, ZM and Stick, JA (2004) In vitro effects of oxytetracycline on matrix metalloproteinase-l mRNA expression and on collagen gel contraction by cultured myofibroblasts obtained from the accessory ligament of foals. Am J Vet Res 65:491-496. Arnoczky, SP, Tian, T, Lavagnino, M and Gardner, K (2004) Ex vivo static tensile loading inhibits MMP-l expression in rat tail tendon cells through a cytoskeletally based mechanotransduction mechanism. J Orthop Res 22:328-333. Arnoczky, SP, Tian, T, Lavagnino, M, Gardner, K, Schuler, P and Morse, P (2002) Activation of stress-activated protein kinases (SAPK) in tendon cells following cyclic strain: the effects of strain frequency, strain magnitude, and cytosolic calcium. J Orthop Res 202947-952. Banes, AJ, Horesovsky, G, Larson, C, Tsuzaki, M, J udex, S, Archambault, J, Zernicke, R, Herzog, W, Kelley, S and Miller, L (1999) Mechanical load stimulates expression of novel genes in vivo and in vitro in avian flexor tendon cells. Osteoarthritis Cartilage 7: 141-153. Banes, AJ, Sanderson, M, Biotano, S, Hu, P, Brigman, B, Tsuzaki, M, Fischer, T and Lawrence, WT (1994) Mechanical load :1: growth factors induce [Ca2+]I release, cyclin D1 expression and DNA synthesis in avian tendon cells. In: V Mow, F Guilak, R Tran-Son-Tay and R Hochmuth (Ed.), Cell Mechanics and Cellular Engineering. Springer, New York, pp. Pages. Banes, AJ, Tsuzaki, M, Hu, P, Brigman, B, Brown, T, Almekinders, L, Lawrence, WT and Fischer, T (1995) PDGF-BB, IGF-I and mechanical load stimulate DNA synthesis in avian tendon fibroblasts in vitro. J Biomech 28:1505-1513. Banes, AJ, Tsuzaki, M, Yamamoto, J, Fischer, T, Brigman, B, Brown, T and Miller, L (1995) Mechanoreception at the cellular level: the detection, interpretation, and diversity of responses to mechanical signals. Biochem Cell Biol 73:349-365. 68 Brown, RA, Prajapati, R, McGrouther, DA, Yannas, IV and Eastwood, M (1998) Tensional homeostasis in dermal fibroblasts: mechanical responses to mechanical loading in three-dimensional substrates. J Cell Physiol 175:323-332. Brown, RA, Talas, G, Porter, RA, McGrouther, DA and Eastwood, M (1996) Balanced mechanical forces and microtubule contribution to fibroblast contraction. J Cell Physiol 169:439-447. Chen, CS and Ingber, DE (1999) Tensegrity and mechanoregulation: from skeleton to cytoskeleton. Osteoarthritis Cartilage 7:81-94. Chicurel, ME, Chen, CS and Ingber, DE (1998) Cellular control lies in the balance of forces. Curr Opin Cell Biol 10:232-239. Dickinson, RB, Guido, S and Tranquillo, RT (1994) Biased cell migration of fibroblasts exhibiting contact guidance in oriented collagen gels. Ann Biomed Eng 22:342- 356. Eastwood, M, McGrouther, DA and Brown, RA (1994) A culture force monitor for measurement of contraction forces generated in human dermal fibroblast cultures: evidence for cell-matrix mechanical signalling. Biochim Biophys Acta 1201 : 186- 192. Eastwood, M, McGrouther, DA and Brown, RA (1998) Fibroblast responses to mechanical forces. Proc Inst Mech Eng [H] 212285-92. Eastwood, M, Porter, R, Khan, U, McGrouther, G and Brown, R (1996) Quantitative analysis of collagen gel contractile forces generated by dermal fibroblasts and the relationship to cell morphology. J Cell Physiol 166233-42. Fringer, J and Grinnell, F (2001) Fibroblast quiescence in floating or released collagen matrices: contribution of the ERK signaling pathway and actin cytoskeletal organization. J Biol Chem 276:31047-31052. Frost, HM (1987) Bone "mass" and the "mechanostat": a proposal. Anat Rec 219: 1-9. Goomer, RS, Basava, D and Maris, T (1999) Effect of stress deprivation on MMP-l gene expression and regulation of MMP-1 promoter in medial collateral and anterior cruciate ligaments (MCL, ACL) and patellar tendon (PT ). Transactions of the Orthopaedic Research Society 24:45. Grinnell, F (1994) Fibroblasts, myofibroblasts, and wound contraction. J Cell Biol 124:401-404. Grinnell, F (1999) Signal transduction pathways activated during fibroblast contraction of collagen matrices. Curr Top Pathol 93:61—73. 69 Grinnell, F (2000) Fibroblast-collagen-matrix contraction: growth—factor signalling and mechanical loading. Trends Cell Biol 10:362-365. Grinnell, F, Zhu, M, Carlson, MA and Abrams, JM (1999) Release of mechanical tension triggers apoptosis of human fibroblasts in a model of regressing granulation tissue. Exp Cell Res 248:608-619. Harris, AK, Stopak, D and Wild, P (1981) Fibroblast traction as a mechanism for collagen morphogenesis. Nature 290:249-251. He, Y and Grinnell, F (1994) Stress relaxation of fibroblasts activates a cyclic AMP signaling pathway. J Cell Biol 126:457-464. Ingber, D (1991) Integrins as mechanochemical transducers. Curr Opin Cell Biol 3:841- 848. Ingber, DE (1997) Tensegrity: the architectural basis of cellular mechanotransduction. Annu Rev Physiol 59:575-599. Kolodney, MS and Wysolmerski, RB (1992) Isometric contraction by fibroblasts and endothelial cells in tissue culture: a quantitative study. J Cell Biol 117:73-82. Lambert, CA, Soudant, EP, N usgens, BV and Lapiere, CM (1992) Pretranslational regulation of extracellular matrix macromolecules and collagenase expression in fibroblasts by mechanical forces. Lab Invest 66:444-451. Langholz, O, Rockel, D, Mauch, C, Kozlowska, E, Bank, 1, Krieg, T and Eckes, B (1995) Collagen and collagenase gene expression in three-dimensional collagen lattices are differentially regulated by alpha 1 beta 1 and alpha 2 beta 1 integrins. J Cell Biol 13121903-1915. Lavagnino, M, Arnoczky, SP, Frank, K and Tian, T (2005) Collagen fibril diameter distribution does not reflect changes in the mechanical properties of in vitro stress-deprived tendons. J Biomech 38:69-75. Lavagnino, M, Arnoczky, SP, Tian, T and Vaupel, Z (2003) Effect of amplitude and frequency of cyclic tensile strain on the inhibition of MMP-1 mRNA expression in tendon cells: an in vitro study. Connect Tissue Res 44:181-187. Lee, TL, Lin, YC, Mochitate, K and Grinnell, F (1993) Stress—relaxation of fibroblasts in collagen matrices triggers ectocytosis of plasma membrane vesicles containing actin, annexins H and VI, and beta 1 integrin receptors. J Cell Sci 105 (Pt 1):167- 177. Lin, YC and Grinnell, F (1993) Decreased level of PDGF-stimulated receptor autophosphorylation by fibroblasts in mechanically relaxed collagen matrices. J Cell Biol 122:663-672. 70 Loitz, BJ, Zernicke, RF, Vailas, AC, Kody, MH and Meals, RA (1989) Effects of short- term immobilization versus continuous passive motion on the biomechanical and biochemical properties of the rabbit tendon. Clin Orthop Relat Res 265-271. Mauch, C, Adelmann-Grill, B, Hatamochi, A and Krieg, T (1989) Collagenase gene expression in fibroblasts is regulated by a three-dimensional contact with collagen. FEBS Lett 250:301-305. Mochitate, K, Pawelek, P and Grinnell, F (1991) Stress relaxation of contracted collagen gels: disruption of actin filament bundles, release of cell surface fibronectin, and down—regulation of DNA and protein synthesis. Exp Cell Res 193: 198-207. Pourati, J, Maniotis, A, Spiegel, D, Schaffer, JL, Butler, JP, Fredberg, JJ, Ingber, DE, Stamenovic, D and Wang, N (1998) Is cytoskeletal tension a major determinant of cell deformability in adherent endothelial cells? Am J Physiol 274:C1283-1289. Prajapati, RT, Chavally-Mis, B, Herbage, D, Eastwood, M and Brown, RA (2000) Mechanical loading regulates protease production by fibroblasts in three- dimensional collagen substrates. Wound Repair Regen 82226-237. Ralphs, JR, Waggett, AD and Benjamin, M (2002) Actin stress fibres and cell-cell adhesion molecules in tendons: organisation in vivo and response to mechanical loading of tendon cells in vitro. Matrix Biol 21:67-74. Rosenfeldt, H, Lee, DJ and Grinnell, F (1998) Increased c-fos mRNA expression by human fibroblasts contracting stressed collagen matrices. Mol Cell Biol 18:2659- 2667. Roy, P, Petroll, WM, Cavanagh, HD and Jester, JV (1999) Exertion of tractional force requires the coordinated up-regulation of cell contractility and adhesion. Cell Motil Cytoskeleton 43:23-34. Rubin, C, Sun, YQ, Hadjiargyrou, M and McLeod, K (1999) Increased expression of matrix metalloproteinase-1 in osteocytes precedes bone resorption as stimulated by disuse: evidence for autoregulation of the cell's mechanical environment? J Orthop Res 17:354-361. Sims, JR, Karp, S and Ingber, DE (1992) Altering the cellular mechanical force balance results in integrated changes in cell, cytoskeletal and nuclear shape. J Cell Sci 103 (Pt 4): 1215-1222. Skalak, R, Skierczynski, BA, Usami, S and Chien, S (1994) Mechanics of cell locomotion. In: Cell mechanics and cellular engineering, VC Mow, F Guilak, R Tran-Son-Tay and R Hochmuth (eds.), pp. New York: Springer. Stemlicht, MD and Werb, Z (2001) How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 17:463-516. 71 Stopak, D and Harris, AK (1982) Connective tissue morphogenesis by fibroblast traction. 1. Tissue culture observations. Dev Biol 90:383-398. Takakuda, K and Miyairi, H (1996) Tensile behaviour of fibroblasts cultured in collagen gel. Biomaterials 17: 1393-1397. Tomasek, JJ, Haaksma, CJ, Eddy, RJ and Vaughan, MB (1992) Fibroblast contraction occurs on release of tension in attached collagen lattices: dependency on an organized actin cytoskeleton and serum. Anat Rec 232:359-368. Tsuzaki, M, Bynum, D, Almekinders, L, Yang, X, Faber, J and Banes, AJ (2003) ATP modulates load-inducible IL-lbeta, COX 2, and MMP-3 gene expression in human tendon cells. J Cell Biochem 89:556-562. Tsuzaki, M, Guyton, G, Garrett, W, Archambault, JM, Herzog, W, Almekinders, L, Bynum, D, Yang, X and Banes, AJ (2003) IL-1 beta induces COX2, MMP-l, -3 and -13, ADAMTS-4, IL-1 beta and IL-6 in human tendon cells. J Orthop Res 21:256-264. Unemori, EN and Werb, Z (1986) Reorganization of polymerized actin: a possible trigger for induction of procollagenase in fibroblasts cultured in and on collagen gels. J Cell Biol 103:1021-1031. Werb, Z and Chin, JR (1998) Extracellular matrix remodeling during morphogenesis. Ann N Y Acad Sci 857: 1 10-1 18. 72 CHAPTER 3 Collagen Fibril Diameter Distribution Does Not Reflect Changes in the Mechanical Properties of in vitro Stress-Deprived Tendons Michael Lavagnino Steven P. Arnoczky Katherine Frank2 Tao Tian From the Laboratory for Comparative Orthopaedic Research College of Veterinary Medicine, Michigan State University, East Lansing, Michigan 48824, USA 2Kalamazoo College, Kalamazoo, Michigan 49006, USA Lavagnino, M, Arnoczky, SP, Frank, K and Tian, T (2005) Collagen fibril diameter distribution does not reflect changes in the mechanical properties of in vitro stress- deprived tendons. J Biomech 38:69-75. 73 ABSTRACT The purpose of this study was to determine if an association exists between the tensile properties and the collagen fibril diameter distribution in in vitro stress-deprived rat tail tendons. Rat tail tendons were paired into two groups of 21 day stress-deprived and 0 time controls and compared using transmission electron microscopy (n = 6) to measure collagen fibril diameter distribution and density, and mechanical testing (n =6) to determine ultimate stress and tensile modulus. There was a statistically significant decrease in both ultimate tensile strength (control: 17.95+/-3.99 MPa, stress-deprived: 6.79+/-3.91 MPa) and tensile modulus (control: 312.8+/-89.5 MPa, stress-deprived: 176.0+/-52.7 MPa) in the in vitro stress-deprived tendons compared to controls. However, there was no significant difference between control and stress-deprived tendons in the number of fibrils per tendon counted, mean fibril diameter, mean fibril density, or fibril size distribution. The results of this study demonstrate that the decrease in mechanical properties observed in in vitro stress-deprived rat tail tendons is not correlated with the collagen fibril diameter distribution and, therefore, the collagen fibril diameter distribution does not, by itself, dictate the decrease in mechanical properties observed in in vitro stress-deprived rat tail tendons. 74 INTRODUCTION Tendons are composite aggregations of Type I collagen, elastin, proteoglycans, glycolipids, water, and cells. The collagen fibrils and their hierarchial arrangement are thought to play an important role in the tensile properties of the tendon (Kastelic et a1. 1978; Viidik 1972). Previous research has documented changes in collagen fibril diameter distributions in tendons and ligaments as a result of aging (Derwin and Soslowsky 1999; Parry et a1. 1978), stress (Cherdchutham et al. 2001; Zachos et al. 2002), stress deprivation (Binkley and Peat 1986; Nakagawa et al. 1989), genetically induced alterations in the extracellular matrix (Clark et al. 2001; Derwin and Soslowsky 1999), healing (Christel and Gibbons 1993; Frank et al. 1992), and anterior cruciate ligament graft remodeling (Frogameni et al. 1993; Oakes 1993). Several studies have correlated a decrease in mean collagen fibril diameter with a loss of mechanical properties in healing and remodeling ligaments and tendons (Frogameni et al. 1993; LaPrade et a1. 1997; Oakes 1993). Most authors attribute this to an increase in the production of small diameter collagen fibers associated with an increase in type III collagen synthesis (Shino et a1. 1995). However, a recent in vitro study has suggested that the decrease in mean collagen fibril diameter seen in remodeling tendon grafts may, in part, be a result of enzymatic (collagenase) degradation of endogenous large collagen fibrils (Cunningham et a1. 1999). In that study, rabbit medial collateral ligaments exposed to bacterial collagenase for 72 or 144 hours demonstrated a significant reduction in mean collagen fibril diameter. Interstitial collagenase has been implicated in the loss of material properties associated with immobilization of ligament and tendons (Goomer et a1. 1999; Loitz et al. 75 1989). Several experimental studies have documented an increase in interstitial collagenase mRNA expression following in vivo immobilization or in vitro stress- deprivation of ligaments and tendons (Arnoczky et al. 2004; Lavagnino et al. 2003; Majima et al. 2000). Thus, it is possible that interstitial collagenase may affect the mechanical properties of stress-deprived tendons and ligaments by enzymatically degrading endogenous collagen fibers. The purpose of this study was to determine if an association exists between the tensile properties and the collagen fibril diameter distribution in a previously described in vitro, stress-deprived, rat tail tendon model (Arnoczky et al. 2004). hi this model, in vitro stress deprivation has been shown to result in a significant increase in rat interstitial collagenase (MMP-13) mRNA and protein expression (Arnoczky et a1. 2004). We hypothesized that in vitro stress deprivation for 21 days would result in a decrease in tensile properties (tensile modulus and failure stress) and that this change in material properties would reflect a decrease in mean collagen fibril diameter. MATERIALS AND METHODS Drugs and Chemicals Dulbecco’s modified Eagle medium (DMEM), fetal bovine serum (FBS), Ascorbate, gentamicin, and penicil1in-streptomycin-fungizone solution were obtained from Gibco (Grand Island, New York, USA). Rat Tail Tendons Following institutional animal care and use approval, tendons were obtained for this investigation from the tails of two 13-month-old Sprague-Dawley rats, euthanized with sodium pentobarbital injection. The tendons were removed immediately after 76 euthanasia. Using a sterile scalpel blade, the tail was cut between coccygeal vertebrae at both the base and at the distal tip of the tail for a total length of approximately 120 mm. Tendons were gently teased from the distal portion of each tail with forceps and placed into a petri dish containing DMEM media supplemented with 10% FBS, antibiotic/antimycotic solution and Ascorbate. The rat tail tendons were then cut in half and paired into two groups: stress-deprived and control. The control group was immediately processed for either transmission electron microscopy (n = 6) or mechanical testing (n = 6). The stress-deprived tendon group were maintained in the above media incubated at 37°C and 10% C03 for 21 days, with media changed three times per week, before processing for either transmission electron microscopy (n = 6) or mechanical testing (n = 6). An additional 30 tendons were divided into control (0 time) and stress- deprived (21 days) groups for a Northern blot analysis to assay for MMP-13 mRNA expression. Transmission Electron Microscggy Tendons were placed in 4% paraformaldehyde for 24 hours, transferred to 4% formalin, and then sent to be cut and embedded for transmission electron microscopy (TEM). The tendons were cut in cross section three times, with the location of the cuts equally distributed along the length of the tendon. The cross sections (100 nm thick) of each fixed tendon were rinsed in 0.1M phosphate buffer then placed in 1% osmium tetroxide in 0.1M phosphate buffer for 3 hours. They were then dehydrated in graded ethanol solutions (30%, 50%, 65%, 75%, 95%, 100%), and transferred to propylene oxide. Tendons were then infiltrated with an Epon-type resin (Poly/Bed 812; Araldite, and dodenyl succinic anhydride [DDSA] in ratios of 5:4:12 [Polysciences, Inc, 77 Warrington, PA, USA]). The resin was infiltrated in three steps (50%, 75%, and 100% resin: propylene oxide). Each infiltration process was performed for 12 hours, and then the specimens were hardened at 60°C for 48 hours. Thin sections, three from the center of each rat tail tendon, were stained with aqueous uranyl acetate and lead citrate, and then scanned with a Philips 301 TEM (Philips Electronic Instrument Co. Roselle, IL, USA). Three random fields were photographed from each section at a magnification of X 19,000. Image Analysis All images were scanned using a Hewlett Packard ScanJet Ich (Palo Alto, CA, USA), and then quantitatively analyzed using Scion Image Beta 4.0.2 software (Scion Corporation, Frederick, MD, USA). All fibrils within each scanned field were analyzed. To eliminate any potential error due to non-perpendicular cuts, the collagen fibril minor diameter was used to represent actual collagen fibril diameter. The area of each collagen fibril was measured and used to calculate the collagen fibril density, defined as the sum of the area of collagen fibrils in the image divided by the total image area. Mechanical Testing Tendons were frozen at -80°C in media until testing. At the time of testing, tendons were thawed to room temperature and a 5 mm portion of each tendon dissected and placed in a saline filled 12-well plate to measure initial tendon diameter with a calibrated microscope. The tendon diameter was determined by taking the mean of four measurements perpendicular to the long axis of the tendon and a circular cross section was assumed for the computation of initial area (Haut 1983; Haut 1985; Haut 1986; Rowe 1985). The remaining portion of the tendon was gripped at the ends with saw— tooth clamps for a 40 mm gage length. The portion of the tendon under each clamp was 78 first air-dried and placed between two pieces of emery board while the midsection (test area) was kept moistened with saline (Haut 1983; Haut 1985; Haut 1986). Since dehydration increases the strength of rat tail tendons by nearly 5 times (Betsch and Baer 1980), premature fracture at the grip was eliminated. The gripped tendon was then mounted onto a custom—made material testing system. The system was equipped with a 5 lb load cell (Sensotec, Columbus, OH, USA), a linear variable differential transformer (LVDT) (Lucas Schaevitz, Pennsauken, NJ, USA) to measure grip-to-grip tendon displacement, and a motion controller (Newport, Fountain Valley, CA, USA) to strain the tendons at a constant rate of 0.168 mm/s (~0.42% strain/s). Each tendon was preloaded to 10 g then loaded at the above rate to failure in a phosphate buffered saline (PBS) bath at room temperature. The load and displacement values were recorded using an analog- to-digital computer data acquisition system. Failure stress, tensile modulus, and failure strain were computed from the load-deformation data. RNA Extraction and Northern Blot Analysis A Northern blot analysis was performed to assay for MMP-l3 mRNA expression after 21-days of stress deprivation in 30 tendons (15 control and 15 stress-deprived). Total cellular RNA was isolated from the rat tail tendons in each group (control and stress-deprived) by the acid guanidine thiocyanate-phenol-chloroform procedure (totally RNA kit, Ambion Inc., Austin, TX, USA) and pooled. The RNA samples were subjected to electrophoresis on 1.2% agarose gels containing 0.66M formaldehyde and MOPS, then transferred to a nylon membrane (Pierce Corp, Rockford, IL, USA) for 1 hour in TAE at 300 mA. Following transfer, the membrane was air-dried and UV cross-linked at 10 Joules/cmz. 79 An MMP-13 probe (Lavagnino et al. 2003) and a human GAPDH cDNA control probe (Clontech Laboratories, Inc., Palo Alto, CA, USA) were labeled with biotin using the North2South Direct HRP Labeling and Detection Kit (Pierce Corp, Rockford, IL, USA). The RNA blots were hybridized with labeled probes (lOng/ml hybridization solution) at 55°C for 1 hour. The membrane was then washed with 40 mL (~ 0.5 mL per cm?) 2x SSC/0.1% SDS at 55°C (3 x 5minutes) and washed with 40 mL (~ 0.5 mL per cm2) of 2x SSC at room temperature. Following the wash procedure, the membrane was incubated with chemiluminescent working solution for 5 minutes and exposed to films for 5-10 minutes. Statistical Analysis Paired t—tests were used to determine any significant statistical difference between control and deprived tendons for the mean fibril diameter and density. A three factor ANOVA was used to determine if there were any differences in collagen diameter distribution between control and deprived tendons, with treatment and size as fixed factors and tendon as a random factor. Paired t-tests were also performed to obtain the statistical difference in both the tensile modulus and ultimate failure strength from the mechanical tests. Statistical significance was set at p<0.05 for all tests. RESULTS Image Analysis Fibril distributions for normal rat-tail tendons exhibit a near equal distribution of both small (<100 nm 52.7%) and large (>100 nm 47.3%) fibrils. There was no significant difference between control and stress-deprived tendons in the number of 80 fibrils per tendon counted, mean fibril diameter, or mean fibril density (Figures 3.1-2, Table 3.1). Figure 3.1 Representative transmission electron microscope image of A: control rat tail tendon cross-section and of B: a cross-section of a rat tail tendon stress-deprived for 21 days (x 19,000). Scale bar = 500 nm. 81 0.5 - a? o 4 L Egg 2 1 $1 0 L3: 2 1‘ C t I E 0.3 - on ro E ii . [Deprived : 3.4% . o a g 0.2 § 3 "11‘ o .i‘ 0.1 - 0.0 “"79 7‘ {3M ‘ $3; Ema; Tendon 1 Tendon 2 Tendon 3 Tendon 4 Tendon 5 Tendon 6 Tendon All Figure 3.2 Histogram illustrating collagen fibril density in control and 21 day stress- deprived rat tail tendons for each of the six paired tendons measured and for all the control and stress-deprived tendons combined. There was no significant differences between tendons, p>0.05. Table 3.1 Mean i standard deviation for control and stress—deprived fibril number, mean fibril diameter and mean fibril density. Resulting p-value from paired t-test with significance set at p<0.05. Fibril Number Mean Fibril Mean Fibril Density, Diameter, nm nmzlnm2 Control 2951 :t 247 150.01 :1: 10.9 0.464 :1: 0.016 Stress-Deprived 2404 1 224 153.65 i 7.11 0.473 :t 0.029 p value 0.17 0.45 0.59 82 There was no significant difference in the fibril size distribution between control and deprived tendons within each fibril diameter bin (Figure 3.3). I Control - -- ——- I Deprived Percentage of Total Fibrils (%) g l O O O O O O O O O O 0 to 0 LO 0 LO 0 ID 0 10 O In Lt.) '— 1— N N CO CO V V to ID I I I I I I I I I I (V) v- v- 1- 1- 1- 1- 1- v- 1- 1- 1!) O l!) O ID 0 l0 0 LO 0 v— V- N N ('3 CO V V LO Fibril flameter (nm) Figure 3.3 Histogram illustrating relative frequencies of collagen fibril diameters in control and 21 day stress-deprived rat tail tendons. There was no significant differences between tendons within each bin, p>0.05. Mechanical Testing There was a statistically significant decrease in both ultimate tensile strength (control: 17.95 i 3.99 MPa, stress-deprived: 6.79 i 3.91 MPa) and tensile modulus (control: 312.8 1- 89.5 MPa, stress—deprived: 176.0 3: 52.7 MPa) in the stress-deprived tendons compared to controls. A representative stress-strain curve from both groups is shown in Figure 3.4. A comparison of tensile modulus, failure stress, and failure strain for each pair of tendon samples is listed in Table 3.2. 83 Rat Tail Tendon Failure Testing 25 (0.168 mm / second) 20- , Control [is 15 — ---- Deprived 2 a)" (I) 9 <75 10 — 1 5- O‘i lllll'T'l‘r'lT'I 0.06 0.08 0.10 0.02 0.04 0.00 Strain, mm/mm Representative stress versus strain curves from paired control and 21 day Figure 3.4 stress-deprived rat tail tendons of one fibril. 84 Table 3.2 Comparison of the cross-sectional area, tensile modulus, failure stress, and failure strain of control and stress-deprived rat tail tendons. TCross- sectional area measurements were paired from the same tendon. The control tendon area was used for both groups to calculate failure stress. * significantly different than control specimens, p<0.05. Control Stress-deprived Tendon Silgjrslal Tensile Failure Failure 85:31:28 Tensile Failure Failure ID Area Modulus Stress Strain Area Modulus Stress Strain (m2) (MPa) (MPa) (%) (mmz) (MPa) (MPa) (%) 1 0.078 455 23.46 6.31 0.078 191 2.90 2.59 4 0.195 262 17.85 9.32 0.195 182 9.72 8.15 5 0.160 230 13.99 7.24 0.160 96 1.14 2.61 6 0.201 227 13.39 7.96 0.201 171 10.35 8.12 9 0.129 346 21.47 7.63 0.129 259 9.69 5.05 11 0.114 357 17.56 7.36 0.114 159 6.93 7.37 Avg. 0.146 312.8 17 .95 7.60 0.146 176.0* 6.79* 5.60“ SD 0.048 89.5 3.99 0.99 0.048 52.7 3.91 2.57 Northern Blot The Northern blot gel demonstrated a complete absence of MMP-l3 mRNA expression in the pooled control tendons. However, MMP-13 mRNA expression was clearly demonstrated in the pooled in vitro stress deprived tendons at 21 days (Figure 3.5). Figure 3.5 MMP- 13 ”W GAPDH C C 1 Control 2 Stress-deprived 21 days Northern blot gel illustrating the relative expression of MMP-13 mRNA expression in fresh control tendons (lane 1) and stress—deprived for 21 days (lane 2). GAPDH was used as an internal control. 85 DISCUSSION Stress deprivation has been shown to have deleterious effects on the structural and functional properties of ligaments and tendons both in vivo and in vitro (Gamble et al. 1984; Hannafin et al. 1995; Majima et al. 1994; Noyes 1977). These cell mediated effects of stress deprivation are thought to produce multiple alterations in the biochemical and biomechanical character of these tissues including an increase in interstitial collagenase mRNA expression (Arnoczky et al. 2004; Goomer et a1. 1999), an increase in collagen degradation and synthesis leading to an increase in collagen turnover and a decrease in collagen mass (Amiel et al. 1983), a decrease in proteoglycans and thus water content (Gamble et al. 1984; Woo et al. 1997), a decrease in ultimate failure stress and tensile modulus (Binkley and Peat 1986; Noyes 1977; Woo et al. 1982), and an increase in reducible collagen crosslinks (Akeson et al. 1987). The results of this 21—day in vitro stress deprivation study demonstrate no significant difference in mean collagen diameter, mean collagen density, or collagen fibril diameter distribution between control and in vitro stress-deprived rat tail tendons. Previous 40-day in vivo stress deprivation studies have produced conflicting ultrastructural results. Binkley and Peat (Binkley and Peat 1986) found a significant increase in the proportion of larger fibrils and a significant decrease in the proportion of smaller fibrils in immobilized rat medial collateral ligaments compared with controls. Nakagawa et a1. (Nakagawa et al. 1989) however, found significantly smaller mean collagen fibril diameter using a hindlimb disuse model of the rat Achilles tendon compared with control tendons. The disparity observed between the results of these studies as well as the results of the current study may be due to differences in 86 methodology, a variability in the degree of stress deprivation actually achieved, or differences in the tissues used. It is also possible that the increase in small diameter collagen fibrils observed in one in vivo study (Nakagawa et al. 1989) could actually be due to an anabolic production of new small diameter fibrils associated with tendon remodeling (Christel and Gibbons 1993; Frogameni et al. 1993; Oakes 1993) rather than a collagenase-induced catabolic deconstruction of large diameter fibrils as suggested by others (Cunningham et al. 1999). In remodeling tissues such as tendon grafts, small diameter fibrils are predominant and are thought to represent newly synthesized collagen (Shino et al. 1995). The mechanical results of the current in vitro stress deprivation study agree with those from several in vivo immobilization studies (Binkley and Peat 1986; Noyes 1977; Woo et al. 1982), which demonstrated significant decreases in tensile modulus and ultimate tensile stress following immobilization. The absence of any significant difference in collagen fibril diameter distribution in the presence of significantly altered material properties seen in the present study suggests that collagen fibril diameter distribution does not solely determine the mechanical properties of rat tail tendons. The ultrastructural results of in vivo stress deprivation studies (Binkley and Peat 1986; Nakagawa et a1. 1989) also lend support to this conclusion as they demonstrate that regardless of the collagen fibril diameter distribution (increase or decrease in small diameter collagen fibrils) the mechanical properties of these stress-deprived tissues diminished over time. Although some studies have correlated changes in collagen fibril diameter with changes in mechanical properties of tendons (Parry et al. 1978) and ligaments (Binkley 87 and Peat 1986), other investigations have suggested that collagen fibril diameter distribution alone cannot predict the material and structural properties of tendons (Derwin and Soslowsky 1999; Derwin et al. 2001) and ligaments (Bay et al. 1993). Researchers have shown other factors beside collagen fibril diameter distribution are important for mechanical integrity and strength of the tendon (Clark et al. 2001; Derwin et al. 2001; Elliott et al. 2003; Haut 1985). Proteoglycans help to regulate the extracellular matrix assembly and structure and are thought to play a role in the structure function relation in tendons (Clark et a1. 2001; Derwin et al. 2001; Elliott et a1. 2003). An increased level of collagen crosslinking has been shown to inhibit the rate of collagenase activity (Woessner and Nagase 2002) and appears to increase the tensile modulus and reduce the strain to failure of collagen (Haut 1985; Thompson and Czernuszka 1995). Other factors that may play a role in determining the structure function relationship of tendons are the crimp pattern (Hansen et al. 2002), the length of the collagen fibril (Trotter and Wofsy 1989), the cell volume fraction or other sources of collagen irregularity (Derwin and Soslowsky 1999; Shrive et a1. 1995) as well as the distribution and amount of proteoglycans and type I collagen (Kronick and Sacks 1994). The results of the current study also demonstrate an increase in levels of rat interstitial collagenase mRN A expression following 21 days of in vitro stress deprivation. Other studies have shown increases in interstitial collagenase mRNA expression in tendons after 24 hours (Arnoczky et al. 2004) and 12 weeks (Goomer et a1. 1999) of stress deprivation. A previous study from our lab using the same rat tail tendon model has demonstrated that MMP-13 mRNA expression correlated with MMP-l3 protein synthesis (Arnoczky et al. 2004). Although MMP-13 expression in this study was clearly 88 present after 21 days of stress deprivation, there was no associated change in the collagen fibril diameter. Thus, the effect of cell-produced collagenase on collagen fibril diameter may not be as immediate or as profound as the previously published effects with bacterial collagenase (Cunningham et al. 1999). This is perhaps due to the impurities of other non-specific proteinases found within bacterial collagenase as well as its different mode of action in degrading type I collagen compared to interstitial collagenase (Woessner and Nagase 2002). The failure strain of the normal control tendons in the current study was similar to that in other investigations (Haut 1985). The decrease in failure strain coupled with the decrease in tensile modulus observed following in vitro stress deprivation in the current study suggests a degradation in the properties of the collagen fibril itself rather than an increase in fibril sliding. However, the mechanism of stress deprivation and MMP-13 expression on the ultrastructural components of extracellular matrix and the subsequent correlation to the mechanical properties of stress-deprived rat tail tendons has yet to be determined. Care must always be taken when comparing similarities and differences between in vivo and in vitro models at both the functional and molecular levels (Hart et al. 2002). It is likely that the level of stress deprivation achieved in the current in vitro system is more complete and more precisely duplicated when compared to the immobilization methods employed in other in vivo systems (Binkley and Peat 1986; Majima et al. 2000; Noyes 1977; Woo et al. 1982). While this may limit the ability to precisely compare the biochemical and physiological sequelae of altered stress conditions in the two systems (owing to the inability to precisely determine the levels of stress deprivation provided by 89 the various immobilization methods) we believe the in vitro stress deprivation model used in the current study is appropriate to test the hypothesis that changes in material properties are related to changes in collagen fibril diameter distribution. In summary, the current study demonstrates that the changes in tensile properties observed in in vitro stress-deprived rat tail tendons are not related to changes in the collagen fibril diameter profile of the tissue. Furthermore, the results of the current study also suggest that, contrary to prior speculation (Cunningham et a1. 1999), the presence of endogenous rat interstitial collagenase (MMP-l3) does not produce an increase in small diameter collagen fibrils in these in vitro stress-deprived tissues. While in vitro stress deprivation in rat tail tendons has been shown to be associated with an increase in MMP- 13 mRNA and protein expression and a decrease in tensile properties, the precise relationship between these sequela have yet to be determined. ACKNOWLEDGEMENTS The authors wish to acknowledge the technical expertise of Ralph Common in obtaining the transmission electron microscope images. The authors would also like to acknowledge the help of Zachary Vaupel and Erika Sorge in their help in analyzing the electron microscope images. 90 REFERENCES Akeson, WH, Amiel, D, Abel, MF, Garfin, SR and Woo, SL (1987) Effects of immobilization on joints. Clin Orthop Relat Res 28-37. Amiel, D, Akeson, WH, Harwood, FL and Frank, CB (1983) Stress deprivation effect on metabolic turnover of the medial collateral ligament collagen. A comparison between nine- and l2-week immobilization. Clin Orthop Relat Res 265-270. Arnoczky, SP, Tian, T, Lavagnino, M and Gardner, K (2004) Ex vivo static tensile loading inhibits MMP—l expression in rat tail tendon cells through a cytoskeletally based mechanotransduction mechanism. J Orthop Res 22:328-333. Bay, B, Howell, SM, Evans, B and Patrissi, GA (1993) An analysis of collagen fiber distributions as a predictor of modulus in a sheep anterior cruciate ligament. Transactions of the Orthopaedic Research Society 18 (2):333. Betsch, DF and Baer, E (1980) Structure and mechanical properties of rat tail tendon. Biorheology 17:83-94. Binkley, JM and Peat, M (1986) The effects of immobilization on the ultrastructure and mechanical properties of the medial collateral ligament of rats. Clin Orthop Relat Res 301-308. Cherdchutham, W, Becker, CK, Spek, ER, Voorhout, WF and van Weeren, PR (2001) Effects of exercise on the diameter of collagen fibrils in the central core and periphery of the superficial digital flexor tendon in foals. Am J Vet Res 62: 1563- 1570. Christel, PS and Gibbons, DF (1993) Collagen fiber changes in the exercised, immobilized, or injured anterior cruciate ligament. In: The Anterior Cruciate Ligament: Current and Future Concepts, DW Jackson (eds), pp. 195-208. New York: Raven Press, Ltd. Clark, RT, Johnson, TL, Schalet, BJ, Davis, L, Gaschen, V, Hunziker, EB, Oldberg, A and Mikic, B (2001) GDP-5 deficiency in rrrice leads to disruption of tail tendon form and function. Connect Tissue Res 42:175-186. Cunningham, KD, Musani, F, Hart, DA, Shrive, NG and Frank, CB (1999) Collagenase degradation decreases collagen fibril diameters—-an in vitro study of the rabbit medial collateral ligament. Connect Tissue Res 40:67—74. Derwin, KA and Soslowsky, LJ (1999) A quantitative investigation of structure-function relationships in a tendon fascicle model. J Biomech Eng 121:598-604. Derwin, KA, Soslowsky, LJ, Kimura, JH and Plaas, AH (2001) Proteoglycans and glycosaminoglycan fine structure in the mouse tail tendon fascicle. J Orthop Res 19:269-277. 91 Elliott, DM, Robinson, PS, Gimbel, JA, Sarver, JJ, Abboud, JA, Iozzo, RV and Soslowsky, LJ (2003) Effect of altered matrix proteins on quasilinear viscoelastic properties in transgenic mouse tail tendons. Ann Biomed Eng 31:599-605. Frank, C, McDonald, D, Bray, D, Bray, R, Rangayyan, R, Chimich, D and Shrive, N (1992) Collagen fibril diameters in the healing adult rabbit medial collateral ligament. Connect Tissue Res 27:251-263. Frogameni, AD, Jackson, DW and Simon, TM (1993) Collagen remodeling in ACL reconstruction (goat model). In: J D.W. (Ed.), The Anterior Cruciate Ligament: Current and Future Concepts. Raven Press, Ltd., New York, pp. Pages. Gamble, JG, Edwards, CC and Max, SR (1984) Enzymatic adaptation in ligaments during immobilization. Am J Sports Med 12:221-228. Goomer, RS, Basava, D and Maris, T (1999) Effect of stress deprivation on MMP—l gene expression and regulation of MMP-1 promoter in medial collateral and anterior cruciate ligaments (MCL, ACL) and patellar tendon (PT). Transactions of the Orthopaedic Research Society 24:45. Hannafin, JA, Arnoczky, SP, Hoonjan, A and Torzilli, PA (1995) Effect of stress deprivation and cyclic tensile loading on the material and morphologic properties of canine flexor digitorum profundus tendon: an in vitro study. J Orthop Res 13:907-914. Hansen, KA, Weiss, JA and Barton, J K (2002) Recruitment of tendon crimp with applied tensile strain. J Biomech Eng 124:72-77. Hart, DA, Natsu-ume, T, Sciore, P, Taseski, V, Frank, CB and Shrive, NG (2002) Mechanobiology: similarities and differences between in vivo and in vitro analysis at the functional and molecular levels. Recent research developments in biophysics and biochemistry 2: 153—177. Haut, RC (1983) Age-dependent influence of strain rate on the tensile failure of rat-tail tendon. J Biomech Eng 105:296-299. Haut, RC (1985) The effect of a lathyritic diet on the sensitivity of tendon to strain rate. J Biomech Eng 107: 166- 174. Haut, RC (1986) The influence of specimen length on the tensile failure properties of tendon collagen. J Biomech 19:951-955. Kastelic, J, Galeski, A and Baer, E (1978) The multicomposite structure of tendon. Connect Tissue Res 6: 1 1-23. Kronick, PL and Sacks, MS (1994) Matrix macromolecules that affect the viscoelasticity of calfskin. J Biomech Eng 116:140-145. 92 LaPrade, RF, Hamilton, CD, Montgomery, RD, Wentorf, F and Hawkins, HD (1997) The reharvested central third of the patellar tendon. A histologic and biomechanical analysis. Am J Sports Med 25:779—785. Lavagnino, M, Arnoczky, SP, Tian, T and Vaupel, Z (2003) Effect of amplitude and frequency of cyclic tensile strain on the inhibition of MMP-1 mRNA expression in tendon cells: an in vitro study. Connect Tissue Res 44:181-187. Loitz, BJ, Zernicke, RF, Vailas, AC, Kody, MH and Meals, RA (1989) Effects of short- term immobilization versus continuous passive motion on the biomechanical and biochemical properties of the rabbit tendon. Clin Orthop Relat Res 265-271. Majima, T, Marchuk, LL, Shrive, NG, Frank, CB and Hart, DA (2000) In-vitro cyclic tensile loading of an immobilized and mobilized ligament autograft selectively inhibits mRNA levels for collagenase (MMP-l). J Orthop Sci 5:503-510. Majima, T, Yasuda, K, Yamamoto, N, Kaneda, K and Hayashi, K (1994) Deterioration of mechanical properties of the autograft in controlled stress-shielded augmentation procedures. All experimental study with rabbit patellar tendon. Am J Sports Med 22:821-829. Nakagawa, Y, Totsuka, M, Sato, T, Fukuda, Y and Hirota, K (1989) Effect of disuse on the ultrastructure of the achilles tendon in rats. Eur J Appl Physiol Occup Physiol 59:239-242. Noyes, FR (1977) Functional properties of knee ligaments and alterations induced by immobilization: a correlative biomechanical and histological study in primates. Clin Orthop Relat Res 210-242. Oakes, BW (1993) Collagen ultrastructure in the normal ACL and in ACL graft. In: DW Jackson (Ed.), The Anterior Cruciate Ligament: Current and Future Concepts. Raven Press, Ltd., New York, pp. Pages. Parry, DA, Barnes, GR and Craig, AS (1978) A comparison of the size distribution of collagen fibrils in connective tissues as a function of age and a possible relation between fibril size distribution and mechanical properties. Proc R Soc Lond B Biol Sci 203:305-321. Rowe, RW (1985) The structure of rat tail tendon fascicles. Connect Tissue Res 14:21- 30. Shino, K, Oakes, BW, Horibe, S, Nakata, K and Nakamura, N (1995) Collagen fibril populations in human anterior cruciate ligament allografts. Electron microscopic analysis. Am J Sports Med 23:203-208; discussion 209. Shrive, N, Chimich, D, Marchuk, L, Wilson, J, Brant, R and Frank, C (1995) Soft-tissue "flaws" are associated with the material properties of the healing rabbit medial collateral ligament. J Orthop Res 13:923-929. 93 Thompson, J1 and Czernuszka, JT (1995) The effect of two types of cross-linking on some mechanical properties of collagen. Biomed Mater Eng 5:37-48. Trotter, J A and Wofsy, C (1989) The Length of Collagen Fibrils in Tendon. Transactions of the Orthopaedic Research Society 14:180. Viidik, A (1972) Interdependence between structure and function in collagenous tissues. In: A Viidik and J Vaust (Ed.), Biology of Collagen. Academic Press, New York, pp. Pages. Woessner, JF and Nagase, H (2002) Matrix Metalloproteinases and TIMPs. In: (Ed.), Oxford University Press, Oxford, pp. Pages. Woo, SL, Gomez, MA, Woo, YK and Akeson, WH (1982) Mechanical properties of tendons and ligaments. II. The relationships of immobilization and exercise on tissue remodeling. Biorheology 19:397-408. Woo, SL, Livesay, GA, Runco, TJ and Young, EP (1997) Structure and function of tendons and ligaments. In: VC Mow and WC Hayes (Ed.), Basic Orthopaedic Biomechanics. Lippincott-Raven, Philadelphia, pp. Pages. Zachos, TA, Arnoczky, SP, Lavagnino, M and Tashman, S (2002) The effect of cranial cruciate ligament insufficiency on caudal cruciate ligament morphology: An experimental study in dogs. Vet Surg 31:596-603. 94 CHAPTER 4 Isolated Fibrillar Damage in Tendons Stimulates Local Collagenase mRNA Expression and Protein Synthesis Michael Lavagnino Steven P. Arnoczky Monika Egerbacher Keri L. Gardner Meghan E. Burns From the Laboratory for Comparative Orthopaedic Research College of Veterinary Medicine, Michigan State University, East Lansing, Michigan 48824, USA Lavagnino, M, Arnoczky, SP, Egerbacher, M, Gardner, KL and Burns, ME (2006) Isolated fibrillar damage in tendons stimulates local collagenase mRNA expression and protein synthesis. J Biomech 39:2355—2362. 95 ABSTRACT The etiology of repetitive stress injuries in tendons has not been clearly identified. While minor trauma has been implicated as an inciting factor, the precise magnitude and structural level of tissue injury that initiates this degenerative cascade has not been determined. The purpose of this study was to determine if isolated tendon fibril damage could initiate an upregulation of interstitial collagenase (MMP13) mRNA and protein in tendon cells associated with the injured fibril(s). Rat tail tendon fascicles were subjected to in vitro tensile loading until isolated fibrillar damage was documented. Once fibrillar damage occurred, the tendons were immediately unloaded to 100g and maintained at that displacement for 24h under tissue culture conditions. In addition, non—injured tendon fascicles were maintained under unloaded (stress-deprived) conditions in culture for 24h to act as positive controls. In situ hybridization or immunohistochemistry was then performed to localize collagenase mRNA expression or protein synthesis, respectively. Fibrillar damage occurred at a similar stress (41.13+/—5.94MPa) and strain (13.24+/- 1.94%) in the experimental tendons. In situ hybridization and immunohistochemistry demonstrated an upregulation of interstitial collagenase mRN A and protein, respectively, in only those cells associated with the damaged fibril(s). In the control (stress-deprived) specimens, collagenase mRNA expression and protein synthesis were observed throughout the fascicle. The results suggest that isolated fibrillar damage and the resultant upregulation of collagenase mRNA and protein in this damaged area occurs through a mechanobiological understimulation of tendon cells. This collagenase production may weaken the tendon and put more of the extracellular matrix at risk for further damage during subsequent loading. 96 INTRODUCTION It has been theorized that overuse injuries in tendons are caused by a failure of the tissue to adapt to repetitive microtrauma and a resultant deterioration of the extracellular matrix over time (Archambault et a1. 1995). However, the precise magnitude and structural level of tissue injury that is required to initiate this degenerative cascade has not been determined. While some investigators have implicated overstimulation of tendon cells as a mechanobiological stimulus for inflammatory cytokine production and matrix degradation (Almekinders et al. 1993; Archambault et al. 2002; Tsuzaki et al. 2003)((Bhargava et al. 2004; Wang et al. 2003) these studies have been performed in monolayer culture and often include high strain magnitudes (Almekinders et a1. 1993; Bhargava et al. 2004; Wang et al. 2003), long loading histories (Archambault et al. 2002; Bhargava et al. 2004; Wang et al. 2003), and the addition of biochemical factors (Archambault et al. 2002; Tsuzaki et al. 2003). Thus, the clinical relevance of such studies is unclear. Recent investigations have demonstrated that fibroblasts are capable of establishing a cytoskeletal tensional homeostasis through interactions with their local extracellular environment (Brown et al. 1998; Lavagnino and Arnoczky 2005). This internal cellular tension has been shown to regulate gene expression in tendon cells and establish the cell’s “calibration point” (Lavagnino and Arnoczky 2005). Mechanical forces which exert additional tension (above and beyond this homeostatic calibration point) to the cytoskeleton will elicit an anabolic response gene, while an absence of mechanical stimuli (or a decrease below the homeostatic level) will elicit a catabolic gene response (Arnoczky et al. 2004; Lavagnino and Arnoczky 2005; Lavagnino et al. 2003). 97 A decrease in extracellular strain in tendons (below homeostatic levels) has been associated with an increase in the upregulation of interstitial collagenase and a subsequent decrease in the tensile properties of these tissues (Arnoczky et al. 2004; Lavagnino et al. 2005). Therefore, it is possible that mechanobiological understimulation of tendon cells, due to an alteration in cell-matrix interactions, could also be an inciting factor in the etiology of overuse injuries. Previous biomechanical studies have suggested that isolated collagen fibril damage occurs near the end of the linear portion of the load deformation curves of ligaments and tendons (Kastelic et al. 1980; Viidik 1972; Woo et al. 1982). While this damage may not affect the ultimate tensile strength of the tissues (Panjabi et al. 1996) it could alter the cell matrix interactions within the damaged portion of the tendon. The alteration of cell matrix interactions secondary to isolated fibrillar damage could cause a mechanobiological understimulation of tendon cells which has been shown to result in an up-regulation of collagenase mRNA expression and protein synthesis (Lavagnino and Arnoczky 2005). This, in turn, could weaken the tendon and put more of the extracellular matrix at risk for further damage with subsequent loading (Lavagnino et al. 2005). A recent study found that a general pattern of collagen deterioration and tissue degeneration was common to both ruptured and tendinopathic tendons suggesting a common, but as yet unidentified, cell mediated, pathological mechanism acting on both of these tendon populations (Tallon et a1. 2001). The purpose of the current study was to induce an isolated fibrillar failure in an in vitro rat tail tendon model system and determine if isolated fibrillar failure results in an up-regulation of collagenase mRNA expression and protein synthesis. The hypothesis 98 was that a mechanobiological understimulation of tendon cells secondary to isolated fibrillar damage in rat tail tendons would result in a local up—regulation in interstitial collagenase mRNA expression and protein synthesis in these cells. MATERIALS AND METHODS Rat Tail Tendon Following institutional animal care and use approval, tendon fascicles were obtained from the tail tendons of adult Sprague-Dawley rats. The fascicles were removed immediately after euthanasia. Using a sterile scalpel blade, the tail was cut between coccygeal vertebrae at both the base and at the distal tip of the tail. Tendon fascicles were gently teased from the distal portion of each tail with forceps and then stored for less than four hours in a 100mm culture dish containing Dulbecco’s Modified Eagle medium (DMEM) supplemented with an antibiotic/antimycotic solution until induction of fibrillar damage testing. During the fibrillar damage testing, the tendon fascicles were maintained in a complete media solution of DMEM supplemented with 10% fetal bovine serum, antibiotic/antimycotic solution, and Ascorbate at room temperature. Ten rat tail tendon fascicles from ten different rats were used to create the isolated fibrillar damage. To confirm that collagenase mRNA expression and protein synthesis was due to fibrillar damage and a resulting lack of mechanobiological stimulus, additional rat tail tendon fascicles were either processed immediately after harvest as negative controls (n=10) or maintained under non-loaded (stress-deprived) conditions in tissue culture for 24 hours as positive controls (n=10). 99 Induction of F ibrillar Damage At the time of testing, a single rat tail tendon fascicle was mounted with a small amount of slack in a custom-designed, low-load, tensile testing apparatus (Arnoczky et al. 2002). The device was equipped with a 5 lb load cell (Sensotec, Columbus, OH, USA) and a linear variable differential transducer (LVDT) (Lucas Schaevitz, Pennsauken, NJ, USA) to measure grip-to-grip tendon displacement. Observations and fascicle diameter (268 i 72 um) measurements were made using a stereomicroscope (Olympus, Melville, NY, USA) with a 7x objective and recorded through the microscope using a CCD camera (Javelin Systems, Torrance, CA, USA). The initial fascicle length (40.4 :I; 0.8 mm) was established by consecutively applying a known displacement (0.001 mm) through a computer controlled stepper motor until the slack was removed and the crimp pattern just began to diminish. The tendon fascicles were loaded starting from this crimped state (0% strain, 0 MPa) at a displacement rate of 20 urn/s until fibrillar damage occurred. Fibrillar damage in the fascicle was determined by visible fibril sliding and a change in the reflectivity of the damaged fibrils as a “crimp” pattern returned to these lax fibrils (Hansen et al. 2002; Kastelic et al. 1978; Viidik and Ekholm 1968). Previous studies have shown that reflective light can be used to delineate loaded (taut) from unloaded (lax) collagen fibers (Hansen et al. 2002; Kastelic et a1. 1978; Viidik and Ekholm 1968). The alteration in reflectivity (and the associated reappearance of crimp) was used to delineate the location and extent of the damaged fibrils. Additionally, fibrillar damage was determined to occur when increases in strain produced a decrease in stress on the recorded stress-strain curve (Figure 4.1). Only those tendons in which isolated fibrillar damage could be documented on both imaging and the stress strain curve 100 were utilized. Immediately following visible fibrillar damage, the tendon fascicles were unloaded to 100 g (Figure 4.1) and maintained at that displacement while incubated at 37°C and 10% C02 for 24 hours. Following this 24 hour incubation period, the tendons were fixed in 4% paraformaldehyde at 4°C until processed for in situ hybridization or immunohistochemistry. 50 .‘(C 40 03 O llllllllllml N 0 Stress (MPa) 03 \ 1O lllLllllllll O 1 Cr T T I T f l l I an f l 0.05 0.1 0.15 0 Strain (mm/mm) Figure 4.1 Representative stress-strain curve of a rat tail tendon fascicle (dark solid line) demonstrating the point at which fibrillar damage occurred (point C), and the unloading of the tendon to 100g (dashed line). A representative curve of the tendon loaded to failure displaying the negative slope in the stress strain curve that eventually ends in total tendon failure (Lavagnino et al. 2005) has also been included (light solid line). Points A-D on the stress-strain curve correspond to the images of the fascicle at those points in Figure 4.2A-D. 101 In situ hybridization To determine the location of cells expressing rat interstitial collagenase (MMP- 13), in situ hybridization was performed on the fixed tendon fascicles (5 injured, 5 fresh, 5 stress—deprived) to identify collagenase mRNA activity. The rat tail tendon fascicle was cut into 15 mm pieces to localize the injury site and reduce the tissue size to facilitate processing. The tendon fascicle segments were incubated in the following solutions: 0.2 N HCl (20 minutes), 3% hydrogen peroxide (20 minutes), RNase-free proteinase K (10 uL/mL for 10 minutes at 37°C) and 4% formalin in PBS. Subsequently, acetylation was performed with acetic anhydride in 0.1 M triethanolamine. The segments were rinsed in 2x SSC (1x SSC containing 150 mM NaCl and 15 mM sodium citrate) at 37°C. For hybridization, antisense and sense probes for rat interstitial collagenase were diluted in hybridization solution (50% deionized formamide, 2x SSC, 50lrg/ml yeast RNA, SmM EDTA, 0.2% Tween 20, 0.5% CHAPS and 100ug/ml Heparin) to a concentration of 2.5llg/mL, and incubated at 90°C for 3 minutes, then on ice for 5 minutes. The prehybridization was carried out at 60°C for 1 hour. The hybridization solution was then layered onto the fascicles and hybridized overnight at 37°C in a humid chamber. Posthybridization washes were performed at 60°C for 3x 15 rrrinutes in 2x SSC with 50% formamide and 10% Chaps, then washed with 2x SSC for 2 minutes. The segments were incubated with block solution (Roche Diagnostics) for 1 hour at room temperature. The digoxigenin-labeled hybrids were detected by antibody incubation performed according to the manufacturer’s instructions (Roche Diagnostics) with the following modifications. A 1:10 dilution of anti-digoxigenin (Fab) conjugated to Rhodamine was used for overnight incubation at room temperature. Then an extra washing step of 0.025% Tween 102 in Tris-buffered saline (pH 7.5) was performed. The fascicle segments were wet mounted on a glass slide for viewing with an Axiovert 200M fluorescent microscope (Carl Zeiss, Thornwood, NY, USA). Preparation of RNA probe A 1167-bp rat interstitial collagenase cDNA fragment was cloned into the pZErOTM-2 vector (Invitrogen Life Technologies). The construct was then linearized by cutting it with EcoRI. In vitro transcription was performed using the appropriate RNA polymerases (T7 RNA polymerase for the anti-sense probe and Sp6 RNA polymerase for the sense probe) in the presence of digoxigenin (DIG)-linked UTP in the reaction mixture (DIG RNA Labeling Kit, Roche Diagnostics). Immunohistochemistry The rat tail tendon fascicles (5 injured, 5 fresh, 5 stress-deprived) were fixed in 4% paraformaldehyde for 24 hours, and rinsed in 70% ethanol and distilled water. The fixed tissues were permeabilized in 0.2% Triton X-100 for 1 hour and antigen retrieval was performed with an unmasking solution (H-3300, VECTOR Lab, Burlingame, CA, USA) for 1 hour at 65°C followed by cooling at room temperature for 20 minutes. Unspecific protein binding was blocked with a solution consisting of 2% horse serum, 1% BSA, 0.1% Triton X-100, and 0.05% Tween-20 in PBS for 1 hour. Anti- MMP-13 (Ab-5, NeoMarkers, Labvision Corp, Fremont, CA, USA) was applied to the fascicles overnight at 4°C at a dilution of 1:80. The primary antibody was detected with anti-rabbit Texas red (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) at a dilution of 1:300. Tendon fascicles were mounted in Vectashield® mounting solution with DAPI (H-1200, 103 VECTOR Lab) and viewed under transmitted and fluorescent light on a Zeiss Axioplan microscope. Statistical Analysis The association between fresh control, injured, and stress-deprived rat tail tendon fascicles and interstitial collagenase gene expression and protein synthesis was evaluated for significance using a minimum chi-square test. Minimum chi-square values were calculated for both gene expression and protein synthesis among the three groups with a critical chi-square value of 5.99 (p=0.05) for significance. RESULTS The crimp pattern on the rat tail tendon fascicles, apparent at 0% strain (Figure 4.2A), disappeared with increasing tension (Figure 4.2B). Fibrillar damage was manifested as a visible sliding of fibrils and a resultant change in the reflectivity of the damaged fibrils (Figure 4.2C). This also coincided with a sudden decrease in stress on the stress-strain curve (Figure 4.1C). The precise location of the isolated fibrillar damage varied between samples. Fibrillar damage occurred at similar stress (41.13 i 5.94 MPa) and strain (13.24 i 1.94 %) values in all experimental tendons. This fibrillar damage occurred at approximately 79% of the failure strain of rat tail tendon fascicles loaded to failure in a previous study (Lavagnino et al. 2005). Upon unloading the tendon fascicles to 100 grams the crimp pattern reappeared within the area of fibrillar injury, but not in the remaining, uninjured, portion of the fascicles (Figure 4.2D). Isolated fibrillar damage did not appear to affect the ability of the remaining intact fascicle to support the 100g tensile loads. Thus, while fibrils within the tendon fascicle were damaged, the fascicle remained structurally intact. 104 Figure 4.2 Images of a rat tail tendon fascicle at various points throughout the testing protocol: A) Prior to loading (the crimp pattern is clearly visible). B) During loading in the linear portion of the stress-strain curve demonstrating the elimination of the crimp pattern. C) Onset of fibrillar damage as manifested by a change in the reflectivity of the damaged fibrils (arrows). D) Unloading of the tendon to 100g and the reoccurrence of the crimp pattern within the damaged fibrils (arrows). (bar = 200 microns) In situ hybridization of all the injured rat tail tendons indicated an up-regulation of MMP-13 mRNA in the cells within the damaged fibril(s) (Figure 4.3). Cells within the remaining, undamaged portion of the tendon fascicle showed no evidence of collagenase mRNA expression. Fresh control fascicles showed no evidence of collagenase mRNA expression while in the unloaded (stress-deprived) control samples, cells throughout the entire fascicle were positive for collagenase mRNA expression (Figure 4.4). 105 100 um Figure 4.3 Representative images of a rat tail tendon fascicle following fibrillar damage. A) The presence of the crimp pattern on the bottom of the tendon fascicle (arrows) indicates the site of isolated fibrillar damage. B) In situ hybridization of the tendon fascicle reveals interstitial collagenase mRNA expression in those cells associated with the damaged fibril(s). The borders of the tendon fascicle are delineated by broken lines. (bar = 100 microns) 100 um Figure 4.4 Representative image of a control (unloaded [stress deprived] for 24 hours) rat tail tendon fascicle demonstrating interstitial collagenase mRNA expression by cells throughout the entire fascicle. (bar = lOOmicrons) Irnmunohistochemical staining for interstitial collagenase protein produced results similar to that seen for collagenase mRNA. Cells within the damaged fibrils stained positive for collagenase protein while cells in the undamaged portion of the tendon fascicle were negative (Figure 4.5). Fresh control fascicles showed no evidence of collagenase protein synthesis while in the unloaded (stress-deprived) control samples, cells throughout the entire fascicle were positive for collagenase protein synthesis. 106 d injured uninjured 1:": up injured uninjure pa Figure 4.5 A) Representative photomicrograph of an injured rat tail tendon fascicle showing the damaged fibrils (denoted by the presence of crimp) immediately adjacent to uninjured fibrils. (bar = 20 microns) B) Photomicrograph of the same field under fluorescent light demonstrating the positive (light gray) staining of MMP-l3 protein in the cytoplasm of only those cells within the damaged fibrils. The nuclei have been counterstained with DAPI (white) to help identify the cells. (bar = 20 microns) A significant association existed between control, injured, and stress deprived tendons and interstitial collagenase mRNA expression (Min. x2 = 14.14, p=0.001) and collagenase protein synthesis (Min. x2 = 14.14, p=0.001). The absence of collagenase mRNA expression and protein synthesis in the fresh control fascicles suggests that the process of removing the rat tail tendon fascicles from the animal did not, in itself, contribute to the increase in collagenase gene expression and protein synthesis. DISCUSSION It is well known that increasing tensile loading alters the structure of tendons through the progressive loss of collagen crimp and the increase in fibril recruitment 107 (Diamant et al. 1972; Hansen et al. 2002; Kastelic et al. 1980; Viidik 1972). At the extremes of physiologic loading, fibril sliding and fibrillar damage occur prior to complete structural failure of the tissue (Kastelic et al. 1980; Viidik 1972; Woo et al. 1982). In the current study, fibrillar damage occurred at a similar strain (13.24 + 1.94%) in all experimental tendons and was documented by a sudden decrease in load as seen on the load deformation curve. This is similar to a recent study which documented fibril sliding in rat tail tendons using confocal laser microscopy (Screen et al. 2004). In that study, the initiation of fibril sliding and damage took place at a median value of 13% strain and was found to coincide with a decrease in tensile stress (Screen et al. 2004). The strain at which fibrillar damage occurred in the current study was approximately 80% of failure strain previously reported for rat tail tendon fascicles (Haut 1985; Lavagnino et a1. 2005). Studies in ligaments have shown that while loading the tissue to 80% of the failure deformation did cause isolated fibrillar damage, this isolated fibrillar damage had no effect on the mechanical properties above 80% deformation including failure load and failure deformation (Panjabi and Courtney 2001; Panjabi et al. 1999; Panjabi et a1. 1996). The investigators suggested that the isolated injury could be sufficient to stimulate a biological response without sacrificing the gross mechanical behavior of the ligament (Panjabi and Courtney 2001). The ability to produce isolated fibril failure within an otherwise intact tendon fascicle may be attributable to the multicomposite structure of the tissue (Kastelic et al. 1980; Viidik 1980). The sequential straightening and loading of crimped collagen fibrils, as well as interfibrillar sliding and shear between fibers and/or fibrils, produce a non- linear load-deformation behavior of tendons that may put certain fibrils “at risk” for 108 damage before others (Viidik 1980; Viidik 1990). Indeed, previous studies have demonstrated that individual tendon fiber and fibril failure begins to occur near the end of the linear range of the stress strain curve prior to complete failure (Viidik 1980; Viidik 1990). While the isolated fibril damage seen in the current study still permitted the remaining tendon to withstand loading (100g) within the linear range of the load deformation curve of the fascicles, the mode of fibril injury or the effect of this injury on the overall mechanical properties of the fascicles was not determined. Further studies are needed to elucidate these micro-injury mechanisms and their impact on tendon material and structural properties. A previous study has shown that isolated fibrillar damage in tendons has been followed by a relaxation of the damaged fibrils (Knorzer et al. 1986). This relaxation (or laxity) of the damaged fibrils was identified by a change in the reflectivity of incident light by these lax collagen fibrils when compared to adjacent loaded fibrils (Hansen et al. 2002; Kastelic et al. 1978; Viidik and Ekholm 1968). The sudden change in light reflectivity (and the reappearance of crimp) in isolated collagen fibrils coincided with a decrease in strain on the stress strain curve confirming fibril damage. In the current study, the reappearance of crimp within the damaged fibril(s) was observed immediately after injury suggesting that these fibrils were no longer transmitting load. The importance of mechanical stress on gene expression in tendon cells has been the subject of several recent investigations (Arnoczky et a1. 2004; Lavagnino and Arnoczky 2005; Lavagnino et al. 2005; Lavagnino et al. 2003). These studies have shown that loss of a homeostatic tensile load on tendon cells results in an immediate upregulation of interstitial collagenase mRNA and protein synthesis 109 (Arnoczky et al. 2004; Lavagnino and Arnoczky 2005 ; Lavagnino et al. 2005; Lavagnino et al. 2003). In the current study, isolated fibrillar damage resulted in the immediate upregulation of collagenase mRNA expression in those tendon cells within the damaged fibril(s). This would suggest an altered cell-matrix interaction within the damaged portion of the fascicle. While this alteration (decrease) in mechanotransduction stimuli is most likely a result of damage to the extracellular matrix (e.g., collagen fibrils, collagen crosslinks, etc.) of the tendon fascicle and a subsequent loss of load-transmitting function, it could also be a result of damage to other components of the mechanotransduction pathway. Fibril sliding in loaded tendons produces strong local shear forces (Knorzer et al. 1986) which could damage cell-matrix adhesions (focal complexes, focal adhesions, or fibrillar adhesions) and/or alter the cytoskeleton. Indeed, a previous study has demonstrated that disruption of the cytoskeleton resulted in an immediate upregulation of collagenase mRNA expression in tendon cells within a loaded tendon (Arnoczky et al. 2004). Additional studies are needed to determine what, if any, portion(s) of the mechanotransduction pathway could be damaged by this fibrillar injury mechanism. Collagenase expression, secondary to mechanobiological understimulation in tendons has been associated with a significant decrease in tensile properties (Lavagnino et al. 2005; Majima et al. 1994). Stress deprivation of rat tail tendons resulted in a significant increase in interstitial collagenase mRNA and protein expression as well as a decrease in both tensile modulus and tensile strength (Lavagnino et al. 2005). A clinical study examining matrix metalloproteinase activity in ruptured human tendons demonstrated a significant increase in interstitial collagenase activity when compared to 110 normal controls (Riley et al. 2002). This increase in collagenase activity was associated with a deterioration in the quality of the collagen network. Another study which examined the histopathology of ruptured and tendinopathic Achilles tendons suggested that while the ruptured tendons were significantly more degenerated than the tendinopathic tendons, the general pattern of tendon degeneration was common to both groups (Tallon et al. 2001). This implies a common pathological mechanism acting on both tendon populations (Tallon et al. 2001) and suggests that tendon degeneration is an active, cell mediated process that may result from a failure to regulate specific matrix- metalloproteinase activities in response to injury (Riley et a1. 2002). The results of the current study demonstrate that isolated fibrillar damage in tendon fascicles results in an upregulation of interstitial collagenase mRNA expression and protein synthesis in those tendon cells associated with the damaged fibril(s). This is thought to occur due to a mechanobiological understimulation of the tendon cells as a result of altered cell-matrix interactions. The upregulation of interstitial collagenase mRNA and protein by tendon cells throughout the unloaded (stress-deprived) control tendon fascicles further supports the role of mechanobiological understimulation of tendon cells in gene expression. The resultant upregulation of interstitial collagenase mRNA expression and protein synthesis may weaken the tendon and put more of the extracellular matrix at risk for further damage with subsequent loading (Lavagnino et al. 2005). Therefore, it is possible that isolated collagen fibrillar damage may have more of an impact on long-term tendon health by altering homeostatic mechanotransduction interactions between the cell and the extracellular matrix rather than from any initial structural weakness of the tendon. lll While any reparative response(s) secondary to the isolated fibrillar damage could not be examined in the in vitro system used in the current study, understanding the nature and extent of such responses would be critical in determining the ability of tendons to recover from this localized fibrillar damage. 112 REFERENCES Almekinders, LC, Banes, AJ and Ballenger, CA (1993) Effects of repetitive motion on human fibroblasts. Med Sci Sports Exerc 25:603-607. Archambault, J, Tsuzaki, M, Herzog, W and Banes, AJ (2002) Stretch and interleukin- 1beta induce matrix metalloproteinases in rabbit tendon cells in vitro. J Orthop Res 20236-39. Archambault, JM, Wiley, JP and Bray, RC (1995) Exercise loading of tendons and the development of overuse injuries. A review of current literature. Sports Med 20:77-89. Arnoczky, SP, Lavagnino, M, Whallon, JH and Hoonjan, A (2002) In situ cell nucleus deformation in tendons under tensile load; a morphological analysis using confocal laser microscopy. J Orthop Res 20229-35. Arnoczky, SP, Tian, T, Lavagnino, M and Gardner, K (2004) Ex vivo static tensile loading inhibits MMP-l expression in rat tail tendon cells through a cytoskeletally based mechanotransduction mechanism. J Orthop Res 22:328-333. Bhargava, M, Attia, ET and Hannafin, JA (2004) The effect of cyclic tensile strain on MMPs, collagen, and casein degrading activities of fibroblasts isolated from anterior cruciate and medial collateral ligaments. Transactions of the Orthopaedic Research Society 50:270. Brown, RA, Prajapati, R, McGrouther, DA, Yannas, IV and Eastwood, M (1998) Tensional homeostasis in dermal fibroblasts: mechanical responses to mechanical loading in three-dimensional substrates. J Cell Physiol 175:323-332. Diamant, J, Keller, A, Baer, E, Litt, M and Arridge, RG (1972) Collagen; ultrastructure and its relation to mechanical properties as a function of ageing. Proc R Soc Lond B Biol Sci 180:293-315. Hansen, KA, Weiss, J A and Barton, J K (2002) Recruitment of tendon crimp with applied tensile strain. J Biomech Eng 124:72-77. Haut, RC (1985) The effect of a lathyritic diet on the sensitivity of tendon to strain rate. J Biomech Eng 107:166-174. Kastelic, J, Galeski, A and Baer, E (1978) The multicomposite structure of tendon. Connect Tissue Res 6:11-23. Kastelic, J, Palley, I and Baer, E (1980) A structural mechanical model for tendon crimping. J Biomech 132887-893. Knorzer, E, Folkhard, W, Geercken, W, Boschert, C, Koch, MH, Hilbert, B, Krahl, H, Mosler, E, Nemetschek-Gansler, H and Nemetschek, T (1986) New aspects of the 113 etiology of tendon rupture. An analysis of time-resolved dynamic-mechanical measurements using synchrotron radiation. Arch Orthop Trauma Surg 105:1 13— 120. Lavagnino, M and Arnoczky, SP (2005) In vitro alterations in cytoskeletal tensional homeostasis control gene expression in tendon cells. J Orthop Res 23:1211-1218. Lavagnino, M, Arnoczky, SP, Frank, K and Tian, T (2005) Collagen fibril diameter distribution does not reflect changes in the mechanical properties of in vitro stress-deprived tendons. J Biomech 38:69-75. Lavagnino, M, Arnoczky, SP, Tian, T and Vaupel, Z (2003) Effect of amplitude and frequency of cyclic tensile strain on the inhibition of MMP-1 mRNA expression in tendon cells: an in vitro study. Connect Tissue Res 44:181-187. Majima, T, Yasuda, K, Yamamoto, N, Kaneda, K and Hayashi, K (1994) Deterioration of mechanical properties of the autograft in controlled stress-shielded augmentation procedures. All experimental study with rabbit patellar tendon. Am J Sports Med 22:821-829. Panjabi, MM and Courtney, TW (2001) High-speed subfailure stretch of rabbit anterior cruciate ligament: changes in elastic, failure and viscoelastic characteristics. Clin Biomech (Bristol, Avon) 16:334-340. Panjabi, MM, Moy, P, Oxland, TR and Cholewicki, J (1999) Subfailure injury affects the relaxation behavior of rabbit ACL. Clin Biomech (Bristol, Avon) 14:24-31. Panjabi, MM, Yoldas, E, Oxland, TR and Crisco, JJ, 3rd (1996) Subfailure injury of the rabbit anterior cruciate ligament. J Orthop Res 14:216-222. Riley, GP, Curry, V, DeGroot, J, van El, B, Verzijl, N, Hazleman, BL and Bank, RA (2002) Matrix metalloproteinase activities and their relationship with collagen remodelling in tendon pathology. Matrix Biol 21:185-195. Screen, HR, Lee, DA, Bader, DL and Shelton, JC (2004) An investigation into the effects of the hierarchical structure of tendon fascicles on micromechanical properties. Proc Inst Mech Eng [H] 218:109-119. Tallon, C, Maffulli, N and Ewen, SW (2001) Ruptured Achilles tendons are significantly more degenerated than tendinopathic tendons. Med Sci Sports Exerc 33:1983- 1990. Tsuzaki, M, Bynum, D, Almekinders, L, Yang, X, Faber, J and Banes, AJ (2003) ATP modulates load-inducible IL-lbeta, COX 2, and MMP-3 gene expression in human tendon cells. J Cell Biochem 89:556—562. 114 Viidik, A (1972) Interdependence between structure and function in collagenous tissues. In: A Viidik and J Vaust (Ed.), Biology of Collagen. Academic Press, New York, pp. Pages. Viidik, A (1980) Mechanical properties of parallel-fibered collagenous tissues. In: Biology of Collagen, A Viidik and J Vuust (eds), pp. 237-255. London: Academic Press. Viidik, A (1990) Structure and function of normal and healing tendon and ligaments. In: Biomechanics of Diarthroidal Joints, VC Mow, A Ratcliffe and SL Woo (eds.), pp. 3-38. New York: Springer. Viidik, A and Ekholm, R (1968) Light and electron microscopic studies of callagen fibers under strain. Z Anat Entwicklungsgesch 127:154-164. Wang, JH, Jia, F, Yang, G, Yang, S, Campbell, BH, Stone, D and Woo, SL (2003) Cyclic mechanical stretching of human tendon fibroblasts increases the production of prostaglandin E2 and levels of cyclooxygenase expression: a novel in vitro model study. Connect Tissue Res 44:128—133. Woo, SL, Gomez, MA, Woo, YK and Akeson, WH (1982) Mechanical properties of tendons and ligaments. II. The relationships of immobilization and exercise on tissue remodeling. Biorheology 19:397-408. 115 CHAPTER 5 A Finite Element Model Predicts the Mechanotransduction Response of Tendon Cells to Cyclic Tensile Loading Michael Lavagninol Steven P. Arnoczkyl Eugene Kepich2 Oscar Caballerol Roger C. Haut2 (1) Laboratory for Comparative Orthopaedic Research, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan 48824, USA (2) Orthopaedic Biomechanics Laboratories, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA Lavagnino, M, Arnoczky, SP, Kepich, E, Caballero, O, Haut, RC (2007) A Finite Element Model Predicts the Mechanotransduction Response of Tendon Cells to Cyclic Tensile Loading. Biomechanics and Modeling in Mechanobiology: Submitted 116 ABSTRACT The importance of fluid—flow-induced shear stress and matrix-induced cell deformation in transmitting the global tendon load into a cellular mechanotransduction response has yet to be determined. A multiscale computational tendon model composed of both matrix and fluid phases was created to examine how global tendon loading may affect fluid-flow-induced shear stresses and membrane strains at the cellular level. The model was then used to develop a quantitative experiment to help better understand the roles of membrane strains and fluid-induced shear stresses on the biological response of individual cells. The model was able to predict the global response of tendon to applied strain (stress, fluid exudation), as well as the associated cellular response of increased fluid-flow-induced shear stress with strain rate and matrix-induced cell deformation with strain amplitude. The model analysis, combined with the experimental results, demonstrated that both strain rate and strain amplitude are able to independently alter rat interstitial collagenase gene expression through increases in fluid-flow-induced shear stress and matrix-induced cell deformation respectively. 117 1 INTRODUCTION Cells are known to respond to physical signals by way of a mechanotransduction tensegrity system, which connects the cell nucleus to the extracellular matrix through a cytoskeleton (Banes et al. 1995; Brown et al. 1998; Ingber 1997). Deformation of the cytoskeleton, via membrane anchored attachment proteins (integrins), or stimulation of other transmembrane proteins (G-protein receptors, receptor kinases, mitogen-activated protein kinases) (Wang 2006) initiates a cascade of gene expressions activating catabolic and/or anabolic cell responses (Lambert et al. 1992; Lavagnino and Arnoczky 2005; Mochitate et a1. 1991). For tendons, the importance of physical stimuli in maintaining homeostasis has been well documented (Hannafin et a1. 1995; Lavagnino et al. 2005; Yasuda and Hayashi 1999). Recent in vitro studies have shown that stress deprivation of tendon cells in situ results in an immediate up-regulation of rat interstitial collagenase mRNA expression (Arnoczky et al. 2004; Lavagnino et al. 2003). Conversely, application of static stress (Arnoczky et al. 2004) or cyclic strain (Lavagnino et a1. 2003) has been shown to inhibit interstitial collagenase mRN A expression in this in vitro tendon model through a cytoskeletally based mechanism. While this gene inhibition has been shown to occur in an amplitude and frequency, dose-dependent manner (Lavagnino et al. 2003), the effect of cyclic loading rate on gene expression has not been explored. Previous studies in bone have demonstrated that both matrix deformation and fluid flow are important regulators of gene expression (Han et al. 2004; Mullender et al. 2004; You et al. 2000). Application of tensile strain to tendons leads to a progressive loss of collagen crimp, an increase in fiber recruitment, and a subsequent deformation of the extracellular matrix (Hansen et al. 2002; Kastelic et al. 1980). Tendon tensile strain has 118 also been shown to modulate tendon cell deformation in a dose-dependent manner (Arnoczky et al. 2002). Cellular deformation has also been implicated as a cell signaling mechanism in tendon cells through a calcium-based pathway (Arnoczky et al. 2007). In addition to matrix deformation, experimental studies have demonstrated interstitial fluid flow in response to cyclic tensile loading of tendons (Hannafin and Arnoczky 1994; Helmer et al. 2006; Lanir et al. 1988). Fluid flow has been shown to control gene expression in a monolayer of tendon cells, presumably through a mechanotransduction mechanism, but without the significant calcium influx seen in deformed cells (Archambault et al. 2002). Therefore, it is possible that the dose- dependent inhibition of collagenase gene expression observed with increasing cyclic load amplitudes and/or frequencies may be related to concomitant increase in deformation and fluid flow induced shear stress on individual cells in the tendon (Lavagnino et a1. 2003). However, the exact levels of fluid-induced shear stress on tendon cells in vivo have not been identified (Archambault et al. 2002). While external loading is known to be an important regulator of bone metabolism, it has been suggested that fluid flow may actually play more of a role than matrix deformation in bone cell mechanotransduction (You et a1. 2000). In tendons, a biphasic model has suggested the importance of distortional strain and hydrostatic stress as important mechanical stimuli regulating the composition of tendon and gene expression (Giori et al. 1993). This and other biphasic tendon models have successfully modeled global tendon properties in response to strain (Atkinson et al. 1997; Giori et al. 1993; Yin and Elliott 2004), but none have predicted local strains or fluid-induced shear stresses on the cell. 119 The mechanical response of tendon to load is determined by its solid (collagen matrix) and fluid components (Atkinson et al. 1997). Collagen fibers and their crimp formation are thought to play a significant role in determining the toe-in region of the nonlinear stress—strain response of tendon (Kastelic et al. 1980). Glycosaminoglycans have also been cited as strong determinants of mechanical properties due to their role in fibril-fibril binding (Redaelli et al. 2003) and their interaction with the fluid in the extracellular matrix (Robinson et a1. 2004). The fluid content of a tendon, or its hydration, is known to alter the mechanical response of tendons to strain rate (Haut and Haut 1997). Although each of these components (extra-cellular matrix deformation and fluid flow) is known to play an important role in the mechanical response of tendon to load, their influence on cellular mechanotransduction is unknown. Therefore, the purpose of this study was to create a multiscale computational tendon model composed of both matrix and fluid phases to examine how global tendon loading may affect stresses and strains at the cellular level. We hypothesized that mechanotransduction signaling in tendon cells can result from either increases in fluid flow induced shear stresses or membrane strains on the tendon cells. The model was developed to study the results of previous experiments (Lavagnino 2003) as well as to guide the development of new experiments to help better understand the roles of membrane strains and fluid-induced shear stresses on the biological response of individual cells. 120 2 MODEL DEVELOPMENT A multi-scale modeling approach was utilized in this study to help predict the levels of fluid-flow-induced shear stress on cells and cell membrane strain generated under various amplitudes and rates of global tendon loading. 2.1 Global Model The global model was based on the geometry and composition of a rat tail tendon (RTT). Tendons are made of three main components: type I collagen (70-80% of dry weight), an extrafibrillar matrix (proteoglycans, glycolipids, cells), and water (60-80% of wet weight) (Woo et al. 1997). Each of these components has mechanically significant functions in the response of tendon to load (Woo et al. 1997). 2.1.1 Collagen Fibers Collagen fibers have been previously modeled as linear elastic (Diamant et al. 1972; Kastelic et al. 1980; Stouffer et al. 1985), bilinear elastic (Belkoff and Haut 1992; Hurschler et a1. 1997; Kwan and Woo 1989), and linear viscoelastic (Johnson et a1. 1996; Lanir 1980; Wang et al. 1997; Wilson et al. 2004). To represent the nonlinear effect of collagen recruitment in the current model, collagen fibers were simulated using axially oriented nonlinear spring elements attached at the nodes of the matrix elements, similar to the model of Wilson et al. (Wilson et al. 1997). The tissue strain at which tendon fibers became uncrimped and began load bearing was assumed to vary linearly in the radial direction, with the largest strain required at the RTT center and the smallest strain at the outer boundary of the RTT (Hansen et a1. 2002). The collagen fiber distribution in tendon was assumed to be uniform with no variation in collagen fiber density. Crosslinks between collagen fibers are formed by secondary collagen fibers (types VI, XII, XIV) 121 and glycosaminoglycans. Although crosslinks are thought to play a role in the mechanical properties of tendon (Haut 1985; Puxkandl et al. 2002; Redaelli et al. 2003; Robinson et al. 2004), they are only indirectly (Poisson’s ratio, low permeability) included in the current model. 2.1.2 Transversely Isotropic Matrix Following the experimental demonstration of fluid exudation from tendon during cyclic tensile loading (Hannafin and Arnoczky 1994; Lanir et al. 1988), computational models have incorporated the effects of permeability and interstitial fluid flow (Adeeb et al. 2004; Atkinson et al. 1997; Butler et a1. 1997; Chen et al. 1998; Yin and Elliott 2004). To adequately model fluid flow in the current model, the RTT matrix was assumed to be transversely isotropic in the fiber or 2-axis direction, with a zero pore pressure enforced on the outer boundary to allow free fluid flow out of the tendon. The transversely isotropic model, with Poisson’s ratio greater than 0.5, has been measured in ligament and shown to be necessary to generate fluid exudation in response to tensile load (Adeeb et al. 2004; Hewitt et a1. 2001; Yin and Elliott 2004). Orthotropic materials may have higher Poisson’s ratios (>0.5) than isotropic materials, as long as thermodynamic constraints are met (Adeeb et al. 2004). A Poisson’s ratio greater than 0.5 is required for volume loss with uniaxial tension, and this volume loss is presumably due to fluid exudation from tendon (Yin and Elliott 2004). The high Poisson’s ratio in tendon may be due to the organization of fibers and their crosslinks and not because of high Poisson’s ratios of the individual tendon components (Adeeb et a1. 2004). The Poisson’s ratio in this model was chosen to reflect the fluid exudation effect of the whole tendon. 122 2.1.3 Tendon Permeability A major determinant of fluid flow is permeability, but the permeability of tendon is not known. However, it has been suggested that permeability is strain (Lai et al. 1981; Weiss and Maakestad 2006; Yin and Elliott 2004), porosity (Chen et al. 1998) and/or voids ratio (van der Voet 1997) dependent. Higher water content in the peripheral rim compared to the tendon core suggests that the porosity or voids ratio may also be depth dependent within a tendon (Wellen et al. 2005). The permeability in tendon is also believed to be anisotropic with a higher permeability along the fiber direction than across the fibers (Butler et al. 1997; Chen et al. 1998; Han et al. 2000). Although the directional dependence within tendon is known, the actual values are still in question. Therefore, for the current model, permeability and voids ratio were assumed to be uniform throughout the tendon. 2.2 Submodel A submodeling function was utilized to provide the output from the global RTT model as boundary conditions in the cell model. The submodel consisted of an ovoid-shaped cell, a low-permeable cell membrane, pericellular and extracellular matrices, and nonlinear collagen fibers. Although tendon cells are often arranged in columns, the current model used a lone cell located on the outer boundary of the tendon. The outer boundary of the tendon was of interest, as cells in this area were assumed to be subject to the largest matrix deformations and fluid flows. In addition, a previous in situ study documented the cell nuclei deformation in this location following applied strain (Arnoczky et al. 2002). The membrane of the cell was assumed to have a low permeability, thus diverting fluid to flow around and not through the cell (Ateshian et al. 2007). Surrounding the cell and cell 123 membrane was the pericellular matrix. The tendon pericellular matrix appears similar in composition to that of cartilage pericellular matrix in that they both contain versican and type VI collagen (Ritty et al. 2003). In cartilage, the pericellular matrix significantly alters the mechanical environment of the cell and thus plays a role in mechanotransduction (Guilak and Mow 2000). In addition, the submodel includes the transversly isotropic extracellular matrix and nonlinear collagen fibers as described for the global model. 3 FINITE ELEMENT METHOD 3.1 Global Model The RTT was assumed to be cylindrically shaped with symmetry along the radial and axial directions. Thus, a global, poroelastic model of the RTT was created 20mm long and 0.15mm in width (Figure 5.1). The RTT was divided into 300, 4-noded, axisymmetric elements (0.2mm x 0.05mm) using the commercial code ABAQUS 6.3 (ABAQUS, Inc. Providence, RI). 3.1.1 Collagen Fibers To represent the nonlinear effect of collagen recruitment, collagen fibers were simulated using axially oriented spring elements attached at the nodes of the matrix elements with a bilinear collagen spring stiffness of ON/mm during fiber straightening and 20N/mm after fiber straightening (Wilson et al. 1997). 124 Figure 5.1 __1\__ if /\ \/ ?>> -_ll_____ll__ /\ /\ <> P>> << > $ I 20mm ll II “4— 0.15mm —> Axisymmetric global poroelastic model of the rat tail tendon (20mm x 0.15mm), divided into 300 4-noded axisymmetric elements (0.2mm x 0.05mm) with radially variant nonlinear spring elements attached at the nodes of the matrix elements (springs), zero pore pressure on the outer boundary (circles), constrained at the tendon center (triangles), and loaded at the tendon end as per previous experimental conditions (arrows). The darkened element boundary indicates the location of the submodel. 125 Total Reaction Force] Outer ------- outer Middle —- - —— — - Center Middle — - — -— - Center 'l I 0.15: / -— 0.1 *- l o 005 w' . ,p' . ,o" ,l' : 4'. 1’ r l ,0" v’ o’ 0‘ o’ o’ 0'. v’ a’ ,o" v’ 4’ ‘f 0’ 1’ I .a. ’7’ ’0’ 0 l .‘M r CAI-4’4 -" -I' r 0 0.5 1 1.5 2 2.5 3 3.5 Strain (%) Reaction Force (N) Figure 5.2 Reaction force (N) plotted against strain (%) to show the radial variation in fiber recruitment from the outer boundary to the inner or center that predicts the nonlinear response of the global tendon. The tendon deformation at which the fiber became uncrimped and began load bearing varied in the radial direction, with the largest strain required at the RTT center and the smallest strain at the outer boundary of the RTT (Figure 5.2) (Hansen et al. 2002). A sensitivity analysis of the global tendon response to each parameter (data not shown) made it apparent that the collagen parameters (stiffness and uncrimping deformation) were dominant determinants of resultant force compared to matrix parameters. Therefore these collagen parameters were manually adjusted to fit the experimental stress-strain curve of a tendon loaded to 3% strain at 6% strain/minute. A constant fiber density of 4 columns of springs was used to mimic all collagen fibers. 126 3.1.2 Transversely Isotropic Matrix To adequately model fluid flow in the current model, the RTT matrix was assumed to be transversely isotropic in the fiber or 2-axis direction. Material properties for the matrix were taken from the range of previous models and experiments on tendon and ligament (Table 5.1) (Adeeb et al. 2004; Atkinson et a1. 1997; Gupta and Haut Donahue 2006; Haridas et al. 1999; Hewitt et al. 2001; Yin and Elliott 2004). Table 5.1 Global matrix material properties. E2 E1=E3 V21=V23 V13=V31 G12=G23 k VOid Parameters (MP3) (MP3) (MP3) (m4/Ns) Ratio Matrix 1.0 0.0457 1.7 0.7 0.1 3.086-14 2.0 Reference 00457.10 00457.10 07.2.73 0.3.09 0157-50 5.5e-18-3.98e-l4 1.0-2.33 Range (Adeeb et al. 2004; Atkinson et al. 1997; Gupta and Haut Donahue 2006; Haridas et al. 1999; Hewitt et a1. 2001; Yin and Elliott 2004) 3.1.3 Boundary Conditions Boundary conditions on the global model included zero pore pressures on the outer boundary to allow free fluid flow out of the tendon (Figure 5.1 circles) and constraint in the fiber direction at the RTT center to account for radial symmetry (Figure 5.1 triangles). The RTT was elongated with an applied displacement to a peak strain in the first cycle at the same applied strain rate as previous experiments: 1% strain at 2% strain per minute (0.017Hz); 1% strain at 20% strain per minute (0.17Hz); 3% strain at 6% strain per minute (0.017Hz) (Figure 5.1 arrows) (Lavagnino et a1. 2003). Additionally the model was also run in the current study at 3% strain and 2% strain per minute (0.0056Hz) to help understand the individual roles of fluid-induced shear stress and membrane strain on interstitial collagenase mRN A expression of tendon cells. 127 3.2 Submodel The submodel was not axisymmetric and was located in the midportion along the length of the global model on the outer boundary (Figure 5.1 darkened element) and composed of 4-node bilinear displacement and pore pressure (CPE4P) elements. 3.2.1 Submodel Components A submodeling function was utilized to provide the output from the global RTT model (displacements, reaction forces, pore pressure) for boundary conditions in the cell model. The submodel was the size of the global element and it consisted of an ovoid-shaped cell, a low-permeable cell membrane, pericellular and extracellular matrices, and nonlinear collagen fibers (Figure 5.3). 7%" T "ix __[., + 200%. 250 . \ 0pm. _L_ \ + ’ 2 —> 10 um. ‘—5O l.lm.—-* Figure 5.3 Submodel of the rat tail tendon, the size of a global element, composed of an ovoid-shaped cell, cell membrane, pericellular matrix (PCM), extracellular matrix (ECM), and collagen fibers. 128 The cell was chosen as an ovoid shape with a major axis length of 24pm and minor axis length of 4pm. The cell membrane in this model was used as a low—permeable barrier between the pericellular matrix and the cell to prevent fluid flow through the cell (Ateshian et a1. 2007). The thickness of the membrane (500nm) was chosen in an attempt to maintain a similar element size as the rest of the submodel and thus maintain numerical stability. This same cell membrane thickness was also used in another submodeling study (Gupta and Haut Donahue, 2006). Two limitations associated with using a thicker membrane than normal (5-10nm) include altered permeability and stiffness values. Having a thicker cell membrane may make the membrane stiffer and thus less deformable. The permeability defined for the cell membrane in this study is higher by a factor of 20 than that previously prescribed to represent the permeability for a 500nm cell membrane (Ateshian et al. 2006). In taking the thickness of the cell membrane into effect we have potentially increased the cell stiffness and permeability and therefore predicted lower cell membrane deformation and fluid flow induced shear stress than if using a normal membrane size (lOnm). Thus, by using a larger membrane thickness to help maintain element stability in the submodel and altering its theoretical permeability, the cell membrane deformations and fluid induced shear stresses were likely altered in an the same direction to preserve the relationship and help validate the overall conclusions of this first study. Some of these issues would necessarily have to be further investigated in future studies to develop this mechanotransduction computational effort. Surrounding the cell and cell membrane is the pericellular matrix with a 2.5um thickness (Gupta and Haut Donahue 2006; Ritty et a1. 2003). Linear poroelastic continuum elements were used to create the cell model with material properties taken 129 from the range of previous models (Table 5.2) (Guilak and Mow 2000; Gupta and Haut Donahue 2006). The transversely isotropic extracellular matrix, as described for the global model, surrounds the pericellular matrix. Nonlinear springs, as collagen fibers, connect only the central and outside edges of the cell model (Figure 5.3). Table 5.2 Submodel material properties Parameter E (MPa) v k (m4/Ns) Void Ratio Pericellular Matrix 1.0 0.490 3.924e-14 2.0 Cell Membrane 1 .0 0.490 4.905e-19 2.0 Cell 0.5 0.069 4.415e-14 2.0 (Guilak and Mow 2000; Gupta and Haut Donahue 2006) 3.2.2 Submodel Analysis Coupled pore fluid diffusion and stress analysis were used to analyze the fluid flow velocities, stresses and strains within the model. Membrane strain was calculated by the change in the perimeter length divided by the original perimeter length as follows: Alf . 8:7. Fluid flow induced shear stress was calculated by the following equation 1' = #331 , where p was the viscosity of the fluid (0.001Pa-s) and gl- was the change in v v the fluid velocity perpendicular to the surface of the cell (Gupta and Haut Donahue 2006). 4 EXPERIMENTAL METHODS To compare the predicted cellular stresses and strains to the interstitial collagenase mRNA inhibition of tendon cells in situ, the following in vitro experiment was performed. 130 4.1 Cyclic Tensile Loading After Institutional Animal Care and Use Committee approval was granted, rat tail tendons were harvested from adult Sprague-Dawley rats immediately after euthanasia. Using a sterile scalpel blade, the tail was cut between coccygeal vertebrae at both the base and at the distal tip of the tail for a total length of approximately 120 mm. Tendons were gently teased from the distal portion of each tail with forceps and maintained in DMEM media supplemented with 10% FBS, antibiotic/antimycotic solution and ascorbate incubated at 37°C and 10% C02. The rat tail tendons were divided into 6 groups as follows: Group A: 1% cyclic strain at 2% strain per minute (0.017Hz) for 24 hours; Group B: 1% cyclic strain at 20% strain per minute (0.17Hz) for 24 hours; Group C: 3% cyclic strain at 6% strain per minute (0.017Hz) for 24 hours; Group D: 3% cyclic strain at 2% strain per minute (0.0056Hz) for 24 hours; Group E: stress-deprived for 24 hours; Group F: fresh (0 hour) control. All experimental input parameters, except Group D, were the same as in previous studies (Lavagnino et al. 2003). Stress-deprived tendons were maintained in a culture dish in complete media under tissue culture conditions. Cyclic strain was applied to tendons in complete media under tissue culture conditions using a custom made, computer controlled, motor driven device (Lavagnino et a1. 2003). While in the previous study Northern blots were used to show alterations in collagenase expression with various global loading parameters, in the current study matrix metalloproteinase (MMP)-l3 (rat interstitial collagenase) mRNA expression was quantified using quantitative real-time polymerase chain reaction. (Q-PCR). 131 4.2 Quantitative Polymerase Chain Reaction At the end of each experimental period, ten tendons from each group were placed in 1.0mL of RNAlaterTM (Qiagen, Valencia, CA) for a period of at least twenty-four hours at 4°C before processing. Total RNA was then extracted using the Qiagen RNEasy Kit (Valencia, CA) with the protocol provided for fibrous tissues. RNA (200-400ng), once concentrations were normalized, was converted into cDNA using the Invitrogen SuperScript III Reverse Transcription system (Carlsbad, CA). Real Time Quantitative PCR was performed using the TaqMan Gene Expression Assay from Applied Biosystems (ABI, Foster City, CA). Samples were run in a 96-well plate (20ltl final volume per reaction) on an ABI 7500-Fast Q-PCR apparatus. The endogenous control used for all Q- PCR experiments was 185 rRNA. Results were analyzed using the Sequence Detection System software available from ABI. TaqMan probe and primer sets were obtained for MMP-l3 (Rn01448197_m1) and 18s rRNA (Hs99999901_sl) from ABI’s Gene Expression Assay database (http://allgenes.com). 5 RESULTS 5.1 Finite Element Results The global model analysis closely predicted the nonlinear stress-strain response of the tendon to 3% strain at 6% strain/minute (Figure 5.4). In addition, the global model also indicated fluid flow in a radial direction leaving the tendon during tensile stretch (Figure 5.5). On the submodel level, this fluid was predicted to flow around the cell with the highest velocity occurring at the cell poles (Figure 5.6). 132 - . Model - - - Experiment Stress, MPa 0 0.5 1 1.5 2 2.5 3 Strain, °/o Figure 5.4 Comparison of tendon model to actual tendon stress-strain response (3% strain at 6% strain/minute). FLVIL. Haqnimd 9 (Ave Crib: 7511 UVWV Figure 5.5 Plot of the fluid velocity magnitude (mm/s) showing fluid flow in the positive direction (arrow) out of the tendon (3% strain at 6% strain/min). The curved lines (springs) represent the collagen fibers. 133 fizz-3H / . jar-- \t\\- -—9—-"—’ I“; 1.— . /. " ¥ \\\ 3%? a a"- em -~ ‘\ \ 33,. ”11‘ . ‘1'. ., T- “V WWW?‘ \ ' \‘ Mme-Less -~ \. \\\ 2;; s33 \ *0 \ “tier; \3‘ \ ‘Vi‘ \‘ \ :2: ".2 -- . \ : 2- 3m.» . s ' \.'\-\ \ - _._., _.:.._ _ , m I} j \ \\ ~ # W... / .. \ \ \ Figure 5.6 Plots of the fluid velocity resultant around the cell and out of the tendon (3% strain at 6% strain/min). The tendon model predicted that increasing both the global strain rate (2% to 6% strain per minute) and strain amplitude (1% to 3%) (group A versus C) would result in an increase in both the maximum shear stress (142%) and cell membrane strain (191%) (Figure 5.7, Table 5.3). With an increase in the applied strain rate from 2% to 20% strain per minute at 1% global model strain (group A versus B), the model predicted a corresponding 124% increase in the maximum localized shear stress at the cell poles, but only a modest (5%) increase in cell membrane strain (Figure 5.7, Table 5.3). In contrast, the theoretical responses of the tendon model for inputs of 1% strain at 20% strain per minute (group B) versus 3% strain at 6% strain per minute (group C) generated similar maximum shear stress values (8% difference). However, the cell membrane strain levels were increased by 175%. The model was then exercised in an attempt to define an experiment to further explore the effects of membrane strain and shear stress. For a 3% strain at 2% strain per rrrinute (group D), the model predicted a cell membrane strain within 8% of group C, with a maximum shear stress that was 145% less than group C (Figure 5.7, Table 5.3). 134 2.5E-03 ——— — I ZOE-03 -——-—— e e 1; I a. II. _ _ _ _ __ _— V : 'I' 0 0/0} ' 'A ‘0 1.5E-03 f "V _n l 1 /o @ 2 , mm 3 . : I 3 1% @ 20%/min -B I; I ‘ '- - - 3% @ 6°/o/min -C I 3 1.0E-03 . — —— - 3% @ 2°/o/min -D 0 I. l L as: 5.05-04 ‘ . ,l m 0.0E+OO é ? . k 3 A: - . . . . . - l 0 30 60 90 120 150 180 210 240 270 300 330 360 angle (deg) Figure 5.7 Graph of shear-stress induced by fluid flow. Note the marked increase in at the polar ends of the cell. Table 5.3 Global model and submodel strain and shear stress values. Group A Group B Group C Group D Global Model Strain, % 1.00 1.00 3.00 3.00 Global Model Strain Rate, % strain/minute 2.00 20.0 6.00 2.00 Cell Membrane Strain. % 1.26 1.33 3.67 3.97 Maximum Shear Stress at 270°. mPa 0.414 0.927 1.004 0.409 Average Shear Stress, mPa 0.157 0.346 0.459 0.179 Collagenase Expression (Lavagnino et al. 2003) Inhibited Eliminated Eliminated NA 5.2 Q-PCR Results The Q-PCR results from each group of tendons suggested direct correlations could be established between the levels of membrane strain and shear stress predicted in the model and the inhibition of collagenase mRNA. These data were calculated relative to the fresh control (0 hours) sample (Figure 5.8). Tendon cells exposed to a higher global strain rate and strain amplitude (group A versus C), expressed a significantly reduced amount of rat interstitial collagenase mRNA (Figure 5.8, Table 5.3). Increasing the global strain rate from 2% to 20% strain per minute at 1% global model strain (group A versus B) also significantly reduced this expression. On the other hand, increasing the amplitude of 135 global strain (1% to 3%), with a reduction of global strain rate from 20% to 6% per minute (group B versus C), had little effect on the expression of rat interstitial collagenase mRNA expression (Figure 5.8, Table 5.3). These data were thus suggestive that enhanced shear stress, rather than increased cell membrane strain was primarily responsible for inhibition of collagenase expression. Furthermore, it was seen that in comparing the results of group C to D, where the fluid induced shear stress on the cell was significantly reduced at 3% strain, the expression of rat interstitial collagenase mRNA was increased. Alternatively, if the Q-PCR results of group A versus D were compared, the collagenase expression was significantly reduced (Figure 5.8). This biological response of the tendon was associated with a theoretical increase in cell membrane strain and little change in the fluid-induced shear stress on the cell (Table 5.3). 3500 2851 3000 7 -7 7- 2500 A r---)“ 2000 -C, , 1000 -7 Relative Quantification of MMP-13 j 33 81 1 54 0 l .r __ J _ Group A Group B Group C Group D Group E Group F 1% strain 1% strain 3% strain 3% strain Stress- Fresh (Oh) (2%/min) (20%/min) (6°/o/min) (2%/min) Deprived Control 1 .__rn Figure 5.8 Gene expression levels of rat interstitial collagenase (MMP-l3) as determined by Real-Time Quantitative PCR. All experimental samples were quantified relative to the fresh (0 hour) control. 136 6 DISCUSSION The results of the initial study showed a strong association between the amount of fluid- flow-induced shear stress on the cell and the inhibition of collagenase mRNA expression, regardless of cell strain. The importance of fluid-flow-induced shear stress was confirmed when the additional boundary condition was predicted and showed that a decrease in shear stress with the same cell membrane strain resulted in less collagenase mRN A inhibition (3% strain at 6% strain per minute [C] versus 3% strain at 2% strain per minute [D]). However, the additional boundary condition also showed that increasing the membrane strain at similar low fluid-induced shear stress also resulted in a decrease in collagenase mRNA expression (1% strain at 2% strain per minute [A] versus 3% strain at 2% strain per minute [D]). Therefore, increased fluid-flow-induced shear stress alone and cell membrane strain alone were shown to inhibit the expression of rat interstitial collagenase mRNA. Thus, the results of this study confirm our hypothesis that fluid- flow-induced shear stresses and matrix-induced cell deformation are able to alter catabolic gene expression, presumably through a mechanotransduction pathway. The interstitial fluid flow and extrusion out of tendon predicted by the model have been documented to occur in tendon following application of static and cyclic tensile loading (Hannafin and Arnoczky 1994; Helmer et al. 2006; Lanir et al. 1988) although the exact amount of fluid exuded per stretch remains unknown. The increased fluid flow predicted around the cell with increased loading rate may explain why collagenase mRNA expression decreases (Lavagnino et al. 2003). Similar to matrix-induced cell deformation, fluid flow has also been suggested to affect cell function and gene expression; however, the method of action has yet to be determined (Hannafin and 137 Arnoczky 1994). A previous study has shown that the fluid flow induced by cyclic or static load did not alter the diffusion of low molecular weight solutes compared to unloaded controls (Hannafin and Arnoczky 1994). These conclusions suggested that any biological benefit of cyclic loading (and resultant fluid flow) is probably not due to an increase in cell nutrition (Hannafin and Arnoczky 1994). Fluid flow, as suggested in bone, may alter gene expression as it induces a drag gradient on the pericellular matrix fibers (Han et al. 2004). The drag gradient on the pericellular fibers, which are connected to the cell membrane, in turn create a drag force on the cell, thereby creating strain amplification from fluid (Han et al. 2004). While the current model does not include pericellular matrix fibers, the increased fluid velocity with increased strain rate suggests that a similar phenomenon could occur in tendons. An increase in fluid-induced shear stress on the cell could also explain the decreased collagenase expression with increased fluid flow. Fluid flow induces shear stress on the cell membrane and can mechanically stimulate cells to alter their gene expression (Archambault et al. 2002; Jin et al. 2000; Ng et al. 2005; You et al. 2000). The current model predicts that cyclic strain at low rate and amplitude results in an average shear stress of 0.157mPa. The magnitude of the shear stress increases significantly with increased rate (0.346mPa) or amplitude and rate (0.459mPa) corresponding with the inhibition of collagenase mRNA expression. While, the amount of fluid-induced shear stress on tendon cells under physiological conditions in vivo is unknown, both cartilage and tendon models have suggested that interstitial fluid flow may be in the range of 54nm/s to lOum/s, resulting in fluid-induced shear of ~65mPa (Ateshian et al. 2007; Frank and Grodzinsky 1987; Ng et al. 2005). In a low flow 138 (63an5) study, myofibroblasts seeded in a 3D collagen gel scaffold were subjected to an estimated average fluid shear stress on the cells that varied between 15 and 33mPa, which were considered superphysiological (Ng et al. 2005). These values of shear stress from interstitial flow have been shown to strongly induce an anabolic response by fibroblasts (Ng et al. 2005). Therefore, the levels of fluid-induced shear stress predicted in this study appear to sufficiently approximate the values needed to inhibit catabolic gene expression. Previous studies have applied no load (stress-deprivation) (Arnoczky et al. 2004; Lavagnino et al. 2003) or much higher levels of fluid-induced shear stress to cells (0.1- 2.5Pa) to induce a catabolic response (Archambault et al. 2002; Jin et al. 2000). Although suggested by the current study, additional studies are needed to determine whether a range of shear stress may exist that result in a homeostatic mechanobiological response, where an imbalance above or below that shear stress range would result in a catabolic response. The matrix-induced cell deformation predicted by the model has also been documented to occur in tendon following application of static tensile load (Arnoczky et a1. 2002; Hansen et al. 2002; Kastelic et a1. 1980). In addition, static tendon load is known to correlate with nuclear strain (Arnoczky et al. 2002) and has been shown to have a dose-dependent effect on gene expression due to cytoskeletal deformation (Arnoczky et al. 2004). Cell deformation and the resulting alterations in the cytoskeleton are key components in the mechanotransduction response(s) of cells (Banes et al. 1995). The dose dependent inhibition of rat interstitial collagenase mRNA with cyclic strain amplitude shown in this study continues to support the mechanosensitive response of cells to deformation. 139 Therefore, both strain rate and strain amplitude appear to play a role in altering cellular gene expression in tendon. This mechanoresponsiveness of tendon cells is vital to maintaining tendon homeostasis (Lavagnino and Arnoczky 2005). The importance of both the fluid and solid components in transmitting mechanical signals to cells has been suggested in other biphasic tissue studies (Guilak and Mow 2000; Gupta and Haut Donahue 2006; You et al. 2000). Although this cellular response to global load has been studied in cartilage and meniscus (Guilak and Mow 2000; Gupta and Haut Donahue 2006), this is the first study to predict the mechanical environment of tendon cells to tensile loading. Although the current study suggests that interstitial collagenase gene expression has both rate and amplitude dose dependence, it was not possible to determine whether rate or amplitude plays a more significant role in maintaining homeostasis. Studies in bone cells suggest fluid flow plays a significantly greater role than substrate deformation in activating gene expression (You et al. 2000). One reason for this dichotomy may be due to how fluid-shear and matrix strains play a role in mechanotransduction. With tendon cell deformation, calcium influx is a first and primary responder prior to alterations in gene expression (Archambault et al. 2002). However, even at high fluid- shear rates and with the induction of collagenase, there is no significant calcium influx in tendon cells (Archambault et al. 2002). Thus, gene expression may be activated through different mechanotransduction mechanisms (kinases, stretch-activated ion channels) depending on the mechanical signal experienced by the cell (Archambault et al. 2002). Indeed, kinases are cell membrane proteins that are phosphorylated when subjected to cyclic stretching or shear stress (Arnoczky et al. 2002; Iwasaki et al. 2000; Wang 2006). 140 A limitation of this study is that permeability is assumed constant throughout the tendon. Previous computational models and experiments have suggested that tendon permeability is anisotropic and strain dependent (Butler et al. 1997; Chen et al. 1998; Han et al. 2000; Yin and Elliott 2004; You et al. 2000). The use of constant permeability may limit the model from determining strain dependent changes in fluid flow that could help explain the weaker rate dependence of interstitial collagenase mRNA expression at higher strain amplitudes seen in this study. Future studies should investigate how the anisotropic, strain-dependent permeability of tendon would affect the magnitude of fluid flow around the cells. Permeability was further investigated over the range of values seen in the literature (data not shown). The results of this analysis showed that the reduced permeability significantly lowered the fluid-flow induced shear stress predicted on the cell, while only modestly lowering the cell membrane strain. However, this change in permeability did not alter the overall relationship of shear stress between groups. The permeability of the PCM would also seem to have a potentially important impact on the shear stress developed on the cell membrane from fluid flow. Thus, this additional analysis demonstrates the importance of determining the material properties of tendon both at the global and cellular level to accurately model tendon mechanical signals on the cell. The current model assumes that the collagen fibers act as springs and therefore the model does not allow for load transfer between fibers or between the fibers and the matrix and cell. Collagen crosslinking with both covalent (Haut 1985) and GAG bonds (Redaelli et al. 2003; Scott 2003) are thought to play an important role in tendon 141 mechanics. A recent study though, does not support the theory that sulfated GAG cross- links influence continuum-level mechanical behavior during quasi-static tensile loading (Lujan et a1. 2007). However, the dermatin sulfate may still contribute to the viscoelastic response when other loading rates or protocols are used (Lujan et al. 2007). The fluid content of tendon determines the strain-rate-sensitive stiffness of tendon (Haut and Haut 1997). Therefore, although the crosslinking effect of GAGs may not be as important as previously theorized, the water content of tendon due to the GAG concentration may still contribute, to both the mechanical properties and the cell response of tendons. Therefore, although the collagen fibers as springs may be able to closely predict the global tendon response, they remain a limitation through their inability to transfer load to the matrix and thus alter the fluid response of the tendon upon loading. The cellular stresses and strains in this study are only evaluated following the global applied displacement to peak strain in the first cycle. Therefore, the correlations between global loading, cell loading and the corresponding gene expression are all based on this one time point. The peak strain of the first cycle was assumed to validly represent the peak values of deformation and shear stress experienced by the cell. However, this assumption is another limitation of this study and additional studies are required to determine how tendon hysterisis and/or stress relaxation may affect mechanical signals at the cellular level over a 24-hour time period of cyclic strain. In conclusion, the model was able to predict the global response of tendon to applied strain (stress, fluid exudation), as well as the associated cellular response of increased fluid-flow-induced shear stress with strain rate and matrix-induced cell deformation with strain amplitude. The model analysis, combined with the experimental 142 results, showed that both strain rate and strain amplitude are able to independently alter catabolic gene expression through increases in fluid-flow-induced shear stress and matrix-induced cell deformation respectively. Although shear stress and cell deformation appear to alter gene expression through mechanotransduction pathways, additional studies are required to separate the importance of these mechanical factors in homeostasis, injury, or rehabilitation. 143 REFERENCES Adeeb, S, Ali, A, Shrive, N, Frank, C and Smith, D (2004) Modelling the behaviour of ligaments: a technical note. Comput Methods Biomech Biomed Engin 7:33-42. Archambault, J M, Elfervig-Wall, MK, Tsuzaki, M, Herzog, W and Banes, AJ (2002) Rabbit tendon cells produce MMP-3 in response to fluid flow without significant calcium transients. J Biomech 35:303-309. Arnoczky, SP, Lavagnino, M and Egerbacher, M (2007) The response of tendon cells to changing loads: Implications in the etiopathogenesis of tendinopathy. In: Tendinopathy in Athletes, SL Woo, P Renstrom and SP Arnoczky (eds), pp. 46- 59. Oxford, England: Blackwell Publishing. Arnoczky, SP, Lavagnino, M, Whallon, J H and Hoonjan, A (2002) In situ cell nucleus deformation in tendons under tensile load; a morphological analysis using confocal laser microscopy. J Orthop Res 20:29-35. Arnoczky, SP, Lavagnino, M, Whallon, J H and Hoonjan, A (2002) In situ cell nucleus deformation in tendons under tensile load; a morphological analysis using confocal laser microscopy. Journal of Orthopaedic Research 20:29-35. Arnoczky, SP, Tian, T, Lavagnino, M and Gardner, K (2004) Ex vivo static tensile loading inhibits MMP-l expression in rat tail tendon cells through a cytoskeletally based mechanotransduction mechanism. J Orthop Res 22:328-333. Arnoczky, SP, Tian, T, Lavagnino, M, Gardner, K, Schuler, P and Morse, P (2002) Activation of stress-activated protein kinases (SAPK) in tendon cells following cyclic strain: the effects of strain frequency, strain magnitude, and cytosolic calcium. J Orthop Res 20:947-952. Ateshian, GA, Costa, KD and Hung, CT (2007) A theoretical analysis of water transport through chondrocytes. Biomech Model Mechanobiol 6291-101. Ateshian, GA, Likhitpanichkul, M and Hung, CT (2006) A mixture theory analysis for passive transport in osmotic loading of cells. J Biomech 39:464-475. Atkinson, TS, Haut, RC and Altiero, NJ (1997) A poroelastic model that predicts some phenomenological responses of ligaments and tendons. J Biomech Eng 1192400- 405. Banes, AJ, Tsuzaki, M, Yamamoto, J, Fischer, T, Brigman, B, Brown, T and Miller, L (1995) Mechanoreception at the cellular level: the detection, interpretation, and diversity of responses to mechanical signals. Biochem Cell Biol 73:349-365. Belkoff, SM and Haut, RC (1992) Microstructurally based model analysis of gamma- irradiated tendon allografts. J Orthop Res 10:461-464. 144 Brown, RA, Prajapati, R, McGrouther, DA, Yannas, IV and Eastwood, M (1998) Tensional homeostasis in dermal fibroblasts: mechanical responses to mechanical loading in three-dimensional substrates. J Cell Physiol 175:323-332. Butler, SL, Kohles, SS, Thielke, RJ, Chen, C and Vanderby, R, Jr. (1997) Interstitial fluid flow in tendons or ligaments: a porous medium finite element simulation. Med Biol Eng Comput 35:742-746. Chen, CT, Malkus, DS and Vanderby, R, Jr. (1998) A fiber matrix model for interstitial fluid flow and permeability in ligaments and tendons. Biorheology 35:103-118. Diamant, J, Keller, A, Baer, E, Litt, M and Arridge, RG (1972) Collagen; ultrastructure and its relation to mechanical properties as a function of ageing. Proc R Soc Lond B Biol Sci 180:293-315. Frank, EH and Grodzinsky, AJ (1987) Cartilage electromechanics--II. A continuum model of cartilage electrokinetics and correlation with experiments. J Biomech 20:629-639. Giori, NJ, Beaupre, GS and Carter, DR (1993) Cellular shape and pressure may mediate mechanical control of tissue composition in tendons. J Orthop Res 11:581-591. Guilak, F and Mow, VC (2000) The mechanical environment of the chondrocyte: a biphasic finite element model of cell-matrix interactions in articular cartilage. J Biomech 33:1663-1673. Gupta, T and Haut Donahue, TL (2006) Role of cell location and morphology in the mechanical environment around meniscal cells. Acta Biomater 2:483-492. Han, S, Gemmell, SJ, Helmer, KG, Grigg, P, Wellen, JW, Hoffman, AH and Sotak, CH (2000) Changes in ADC caused by tensile loading of rabbit achilles tendon: evidence for water transport. J Magn Reson 1442217-227. Han, Y, Cowin, SC, Schaffler, MB and Weinbaum, S (2004) Mechanotransduction and strain amplification in osteocyte cell processes. Proc Natl Acad Sci U S A 101:16689-16694. Hannafin, J A and Arnoczky, SP (1994) Effect of cyclic and static tensile loading on water content and solute diffusion in canine flexor tendons: an in vitro study. J Orthop Res 12:350-356. Hannafin, J A, Arnoczky, SP, Hoonjan, A and Torzilli, PA (1995) Effect of stress deprivation and cyclic tensile loading on the material and morphologic properties of canine flexor digitorum profundus tendon: an in vitro study. J Orthop Res 13:907-914. Hansen, KA, Weiss, J A and Barton, J K (2002) Recruitment of tendon crimp with applied tensile strain. J Biomech Eng 124:72-77. 145 Hansen, KA, Weiss, JA and Barton, J K (2002) Recruitment of tendon crimp with applied tensile strain. Journal of Biomechanical Engineering 124:72-77. Haridas, B, Butler, DL, Malaviya, P, Boivin, G, Awad, H and Smith, F (1999) Transversely Isotropic Poroelastic Finite Element Simulations of the Intact and Surgically Translocated Rabbit Flexor Tendon. ASME Haut, RC (1985) The effect of a lathyritic diet on the sensitivity of tendon to strain rate. J Biomech Eng 107:166-174. Haut, TL and Haut, RC (1997) The state of tissue hydration determines the strain-rate- sensitive stiffness of human patellar tendon. J Biomech 30:79-81. Helmer, KG, Nair, G, Cannella, M and Grigg, P (2006) Water movement in tendon in response to a repeated static tensile load using one-dimensional magnetic resonance imaging. J Biomech Eng 128:733-741. Hewitt, J, Guilak, F, Glisson, R and Vail, TP (2001) Regional material properties of the human hip joint capsule ligaments. J Orthop Res 19:359-364. Hurschler, C, Loitz-Ramage, B and Vanderby, R, Jr. (1997) A structurally based stress- stretch relationship for tendon and ligament. J Biomech Eng 119:392-399. In gber, DE (1997) Tensegrity: the architectural basis of cellular mechanotransduction. Annu Rev Physiol 59:575-599. Iwasaki, H, Eguchi, S, Ueno, H, Marumo, F and Hirata, Y (2000) Mechanical stretch stimulates growth of vascular smooth muscle cells via epidermal growth factor receptor. Am J Physiol Heart Circ Physiol 2782H521-529. J in, G, Sah, RL, Li, YS, Lotz, M, Shyy, J Y and Chien, S (2000) Biomechanical regulation of matrix metalloproteinase-9 in cultured chondrocytes. J Orthop Res 18:899-908. Johnson, GA, Livesay, GA, Woo, SL and Rajagopal, KR (1996) A single integral finite strain viscoelastic model of ligaments and tendons. J Biomech Eng 118:221-226. Kastelic, J, Palley, I and Baer, E (1980) A structural mechanical model for tendon crimping. J Biomech 132887-893. Kwan, MK and Woo, SL (1989) A structural model to describe the nonlinear stress-strain behavior for parallel-fibered collagenous tissues. J Biomech Eng 111:361-363. Lai, WM, Mow, VC and Roth, V (1981) Effects of nonlinear strain-dependent permeability and rate of compression on the stress behavior of articular cartilage. J Biomech Eng 103:61-66. 146 Lambert, CA, Soudant, EP, Nusgens, BV and Lapiere, CM (1992) Pretranslational regulation of extracellular matrix macromolecules and collagenase expression in fibroblasts by mechanical forces. Lab Invest 66:444-451. Lanir, Y (1980) A microstructure model for the rheology of mammalian tendon. J Biomech Eng 102:332-339. Lanir, Y, Salant, EL and Foux, A (1988) Physico-chemical and microstructural changes in collagen fiber bundles following stretch in-vitro. Biorheology 25:591-603. Lavagnino, M and Arnoczky, SP (2005) In vitro alterations in cytoskeletal tensional homeostasis control gene expression in tendon cells. J Orthop Res 23:1211-1218. Lavagnino, M, Arnoczky, SP, Frank, K and Tian, T (2005) Collagen fibril diameter distribution does not reflect changes in the mechanical properties of in vitro stress-deprived tendons. J Biomech 38:69-75. Lavagnino, M, Arnoczky, SP, Tian, T and Vaupel, Z (2003) Effect of amplitude and frequency of cyclic tensile strain on the inhibition of MMP-1 mRN A expression in tendon cells: an in vitro study. Connect Tissue Res 44:181-187. Lujan, TJ, Underwood, CJ, Henninger, HB, Thompson, BM and Weiss, J A (2007) Effect of dermatan sulfate glycosaminoglycans on the quasi-static material properties of the human medial collateral ligament. J Orthop Res Mochitate, K, Pawelek, P and Grinnell, F (1991) Stress relaxation of contracted collagen gels: disruption of actin filament bundles, release of cell surface fibronectin, and down-regulation of DNA and protein synthesis. Exp Cell Res 193:198-207. Mullender, M, El Haj, AJ, Yang, Y, van Duin, MA, Burger, EH and Klein-Nulend, J (2004) Mechanotransduction of bone cells in vitro: mechanobiology of bone tissue. Med Biol Eng Comput 42:14-21. Ng, CP, Hinz, B and Swartz, MA (2005) Interstitial fluid flow induces myofibroblast differentiation and collagen alignment in vitro. J Cell Sci 118:4731-4739. Puxkandl, R, Zizak, 1, Paris, 0, Keckes, J, Tesch, W, Bernstorff, S, Purslow, P and Fratzl, P (2002) Viscoelastic properties of collagen: synchrotron radiation investigations and structural model. Philos Trans R Soc Lond B Biol Sci 357:191-197. Redaelli, A, Vesentini, S, Soncini, M, Vena, P, Mantero, S and Montevecchi, FM (2003) Possible role of decorin glycosaminoglycans in fibril to fibril force transfer in relative mature tendons-~a computational study from molecular to microstructural level. J Biomech 36:1555-1569. Ritty, TM, Roth, R and Heuser, JE (2003) Tendon cell array isolation reveals a previously unknown fibrillin-2-containing macromolecular assembly. Structure (Camb)11:1179-1188. 147 Robinson, PS, Lin, TW, J awad, AF, Iozzo, RV and Soslowsky, LJ (2004) Investigating tendon fascicle structure-function relationships in a transgenic-age mouse model using multiple regression models. Ann Biomed Eng 32:924-931. Scott, JE (2003) Elasticity in extracellular matrix 'shape modules' of tendon, cartilage, etc. A sliding proteoglycan-filament model. J Physiol 553:335-343. Stouffer, DC, Butler, DL and Hosny, D (1985) The relationship between crimp pattern and mechanical response of human patellar tendon-bone units. J Biomech Eng 107:158-165. van der Voet, A (1997) A comparison of finite element codes for the solution of biphasic poroelastic problems. Proceedings of the Institute of Mechanical Engineering 211: Wang, JH (2006) Mechanobiology of tendon. J Biomech 39:1563-1582. Wang, J L, Pamianpour, M, Shirazi-Adl, A and Engin, AE (1997) Failure criterion of collagen fiber: viscoelastic behavior simulated by using load control data. Theory and Application to Fracture Mechanics 27: 1-12. Weiss, J A and Maakestad, BJ (2006) Permeability of human medial collateral ligament in compression transverse to the collagen fiber direction. J Biomech 39:276-283. Wellen, J, Helmer, KG, Grigg, P and Sotak, CH (2005) Spatial characterization of T1 and T2 relaxation times and the water apparent diffusion coefficient in rabbit Achilles tendon subjected to tensile loading. Magn Reson Med 53:535-544. Wilson, A, Shelton, F, Chaput, C, Frank, C, Butler, D and Shrive, N (1997) The shear behaviour of the rabbit medial collateral ligament. Med Eng Phys 19:652-657. Wilson, W, van Donkelaar, CC, van Rietbergen, B, Ito, K and Huiskes, R (2004) Stresses in the local collagen network of articular cartilage: a poroviscoelastic fibril- reinforced finite element study. J Biomech 37:357-366. Woo, SL, Livesay, GA, Runco, TJ and Young, EP (1997) Structure and function of tendons and ligaments. In: Basic Orthopaedic Biomechanics, VC Mow and WC Hayes (eds), pp. 209-251. Philadelphia: Lippincott-Raven. Yasuda, K and Hayashi, K (1999) Changes in biomechanical properties of tendons and ligaments from joint disuse. Osteoarthritis Cartilage 7: 122-129. Yin, L and Elliott, DM (2004) A biphasic and transversely isotropic mechanical model for tendon: application to mouse tail fascicles in uniaxial tension. J Biomech 37:907-916. You, J, Yellowley, CE, Donahue, HJ, Zhang, Y, Chen, Q and Jacobs, CR (2000) Substrate deformation levels associated with routine physical activity are less 148 stimulatory to bone cells relative to loading-induced oscillatory fluid flow. J Biomech Eng 1221387-393. 149 CONCLUSIONS The goal of this dissertation was to investigate the role of cell mechanobiology in tendon health. The response of tendon cells to changing loading conditions has significant implications in unraveling the etiopathogenesis of tendinopathy (Arnoczky IJEP). This dissertation supports the hypothesis that the etiopathogenic stimulus for tendinopathy may occur from the mechanobiologic under-stimulation of tendon cells resulting from altered cell-matrix interactions. Stress-deprivation of rat tail tendons results in the under-stimulation of tendon cells in situ and an immediate up-regulation of interstitial collagenease mRNA expression and protein synthesis [Appendix 1, Chapter 1] (Arnoczky et al. 2004; Lavagnino et al. 2003). Application of cyclic strain to tendons inhibits the up-regulation of this catabolic gene expression in a dose-dependent manner (both amplitude and frequency), presumably through a cytoskeletally based mechanotransduction pathway [Chapter 1] (Lavagnino et al. 2003). The applied strain rate and strain amplitude are able to independently alter interstitial collagenase gene expression through increases in fluid-flow—induced shear stress and matrix-induced cell deformation respectively [Chapter 5]. This ability of cyclic strain to inhibit interstitial collagenase, a degenerative enzyme involved in tendinopathy, may lead to advances in overuse injury prevention and optimal rehabilitation protocols following tendon injury and repair. These results suggest that tendon cells may have a threshold, or set-point, with regard to their sensitivity to tensile loading and the resulting change in gene expression. Further investigation determined that tendon cells, when seeded into collagen gels, were able to demonstrate a mechanostat set point as changes in their cytoskeletal tension 150 (established or lost) corresponded to the alterations of anabolic (collagen) and catabolic (collagenase) gene expression in a reciprocal manner [Chapter 2] (Lavagnino and Arnoczky 2005). The long-term loss of cytoskeletal tension (21-day stress deprivation) in tendons results in a maintained up-regulation of interstitial collagenase, which, in turn, was shown to cause a significant loss of material properties of the tendon [Chapter 3] (Lavagnino et al. 2005). It was also shown that at high strains, isolated fibril damage could occur in tendons where the damaged fibril(s) are no longer able to transmit extracellular matrix loads to the tendon cells [Chapter 4] (Lavagnino et al. 2006). The localized stress-deprivation in this damaged area, will result in an under-stimulation of these cells [Chapter 1] (Lavagnino et a1. 2003) which, initiates a catabolic response that can weaken the tendon [Chapter 3] (Lavagnino et al. 2005) making it more susceptible to damage from subsequent loading [Chapter 4] (Lavagnino et a1. 2006). However the partially damaged tendon fascicle maintained its ability to bear load (Lavagnino et al. 2006). Therefore, it is possible that during a series of repetitive loading cycles a single abnormal loading cycle could produce strains sufficient enough to induce isolated fibril damage, but not cause clinical injury. Thus, while repetitive loading, per se, may not be responsible for initiating the cascade of events that lead to tendinopathy, it is likely that continued loading of the compromised tissue plays a significant role in the progression of the pathological process. Then when a critical level of damage has been reached the clinical and histological signs of tendinopathy may become evident (Arnoczky et al. 2007). Therefore, in conclusion this dissertation suggests that it is actually an absence of mechanical stimuli, secondary to microtrauma that is the mechanobiological stimulus for the degenerative cascade that leads to tendinopathy. 151 REFERENCES Arnoczky, SP, Lavagnino, M and Egerbacher, M (2007) The Mechanobiological Etiopathogenesis of Tendinopathy: Is it the over-stimulation or the under- stimulation of tendon cells? International Journal of Experimental Pathology In Press. Arnoczky, SP, Tian, T, Lavagnino, M and Gardner, K (2004) Ex vivo static tensile loading inhibits MMP-l expression in rat tail tendon cells through a cytoskeletally based mechanotransduction mechanism. J Orthop Res 22:328-333. Lavagnino, M and Arnoczky, SP (2005) In vitro alterations in cytoskeletal tensional homeostasis control gene expression in tendon cells. J Orthop Res 23:1211-1218. Lavagnino, M, Arnoczky, SP, Egerbacher, M, Gardner, KL and Burns, ME (2006) Isolated fibrillar damage in tendons stimulates local collagenase mRNA expression and protein synthesis. J Biomech 39:2355-2362. Lavagnino, M, Arnoczky, SP, Frank, K and Tian, T (2005) Collagen fibril diameter distribution does not reflect changes in the mechanical properties of in vitro stress-deprived tendons. J Biomech 38:69-75. Lavagnino, M, Arnoczky, SP, Tian, T and Vaupel, Z (2003) Effect of amplitude and frequency of cyclic tensile strain on the inhibition of MMP-1 mRNA expression in tendon cells: an in vitro study. Connect Tissue Res 44:181-187. 152 APPENDIX A Ex vivo static tensile loading inhibits MMP-l expression in rat tail tendon cells through a cytoskeletally based mechanotransduction mechanism Steven P. Arnoczky Tao Tian Michael Lavagnino Keri L. Gardner From the Laboratory for Comparative Orthopaedic Research College of Veterinary Medicine, Michigan State University, East Lansing, Michigan 48824, USA Arnoczky, SP, Tian, T, Lavagnino, M, and Gardner, KL (2004) Ex vivo static tensile loading inhibits MMP-l expression in rat tail tendon cells through a cytoskeletally based mechanotransduction mechanism J Ortho Res 22:328-333. 153 ABSTRACT To determine the effect of various degrees of ex vivo static tensile loading on the expression of collagenase (MMP-l) in tendon cells, rat tail tendons were statically loaded in tension at 0.16, 0.77, 1.38 or 2.6 MPa for 24 h. Northern blot analysis was used to assay for mRNA expression of MMP-1 in freshly harvested, 24 h load deprived, and 24 h statically loaded tendons. Western blot analysis was used to assay for pro-MMP-l and MMP-1 protein expression in fresh and 24 h load deprived tendons. Freshly harvested rat tail tendons demonstrated no evidence of MMP-1 mRNA expression and no evidence of the pro-MMP-l or MMP-l protein. Ex vivo load deprivation for 24 h resulted in a marked increase in the mRNA expression of MMP-1 which coincided with a marked increase of both pro-MMP-l and MMP-1 protein expression. When tendons were subjected to ex vivo static tensile loading during the 24 h culture period, a significant inhibition of this upregulation of MMP-1 mRNA expression was found with increasing load (p<0.05). A strong (r2=0.78) and significant (p<0.001) inverse correlation existed between the level of static tensile load and the expression of MMP-1. Disruption of the actin cytoskeleton with cytochalasin D abolished the inhibitory effect of ex vivo static tensile loading on MMP-l expression. The results of this study suggest that up-regulation of MMP-1 expression in tendon cells ex vivo can be inhibited by static tensile loading, presumably through a cytoskeletally based mechanotransduction pathway. 154 INTRODUCTION The deleterious effects of immobilization on the structural and functional properties of ligaments and tendons are well-known [3,11,16,17,26-28]. While these alterations in the extracellular matrix appear to be cell mediated [17], the exact mechanism by which load affects tissue homeostasis is not completely understood. Previous work from our laboratory has demonstrated a progressive and significant decrease in the tensile modulus of load-deprived tendons in an in vitro system [17]. However, this effect was inhibited by application of low level (frequency and magnitude) cyclic strain [17]. Although the exact mechanism by which load deprivation decreases tensile modulus is still unclear, several investigators have implicated the role of interstitial collagenase (MMP-l) in this process [16,27]. A recent study has demonstrated that immobilization-induced up-regulation in the mRNA expression of MMP-1 in rabbit medial collateral ligaments could be inhibited by in vitro tensile loading [27]. Other in vitro studies have suggested that changes in cell shape can be correlated with a program of gene expression which is manifested by the degradation and synthesis of extracellular macromolecules [2,25,32,33]. Indeed, collagenase expression in fibroblasts has been shown to be up-regulated following chemically induced alterations in cell shape and a reorganization of the actin cytoskeleton [2,33]. A recent study has demonstrated that tensile loading of rat tail tendons results in an alteration of cell nuclear shape in a dose dependent manner [5]. Changes in cell and nuclear shape associated with changes in tendon tensile load are thought to be mediated through the extracellular matrix to the nucleus via transmembrane integrins and the cytoskeleton [8,15,31,34,35]. Thus, MMP-l gene expression in tendons and ligament 155 cells could be influenced by mechanical load trhough such “mechanosensory” cytoskeletal tensegrity system(s). Thus, we hypothesized that MMP-l gene expression in tendon cells varies with the magnitude of tensile load applied to the tendon and that this effect is mediated through the cytoskeleton. MATERIALS AND METHODS Tendons were obtained from the tails of adult Sprague Dawley rats. The tendons were removed immediately after euthanasia and maintained in Dulbecco’s modified Eagle medium media supplemented with 10%, FBS, antibiotic/antimycotic solution, and ascorbate at 37 °C and 10% C02 for the duration of the experiments. Tendons were divided into the following groups: Group 1: freshly harvested tendons (time zero); Group 2: load deprived at 0 MPa for 24 h; Group 3: static tensile stress at 0.16 MPa for 24 h: Group 4: static tensile stress at 0.77 MPa for 24 h; Group 5: static tensile stress at 1.38 MPa for 24 h and Group 6: static tensile stress at 2.60 MPa for 24 h. Static tensile load was applied by attaching clips (weighing 1.2 g) alone or clips with individual, calibrated stainless steel weights (5, 10, or 20 g) to an approximately 50 mm length of tendon (Figure 1). Stress values were calculated from representative microscopic measurements of the rat tail tendons (average diameter 320 pm). The weighted tendons were suspended in individual test tubes containing the above described media while load deprived tendons were placed in a 60 mm culture dish with the same media. All specimens were incubated for 24 h at 37 °C and 10% C0; To determine the role of the cytoskeleton on the tensile load-modulated inhibition of MMP-1 mRNA expression, statically loaded (2.6 MPa) rat tail tendons were incubated in media containing 10 uM cytochalasin D (SIGMA, St Louis, MO). 156 Figure A.l Photograph of the static tensile loading system. Individual tendons were suspended in 50 ml test tubes filled with culture media containing 10% FBS and calibrated stainless steel weights were attached by clips. Construction of rat MMP-I3 DNA plasmid: To prepare the probe, rat MMP-l DNA (GeneBank M60616) was amplified by polymerase chain reaction using rat genomic DNA template. The oligonucleotide primers used were 5 ’-GCC CAT ACA GTT TGA ATA CAG TAT CTG-3’ and 5’-CCA GTT TAA TAA ACA CCA TCT CTT GA- 157 3’. PCR product (1167-bp) was subjected to electrophoresis on 1% agarose gel and recovered using Qiaex II Kit (QIAGEN Inc., Valencia. CA), and then cloned into PCR ®2.1-T0P0® plasmid using TOPO TA cloning kit (Invitrogen Living Science, Carlsbad, CA). The plasmid was transformed into competent E. coli with selection for kanamvcin and ampicillin resistance. The construction was confirmed by restriction enzyme digestion (ECOR 1, EcoR V and Hind III) and polymerase chain reaction. RNA extraction und northern blot analysis: Twenty rat tail tendons per group per experiment were used, and the experiment was repeated three times. At the end of the experimental period an approximately 40 mm long segment (which did not include any portion of the tendon in contact with the clips used to attach the weight or to suspend the tendon) of the statically loaded tendons and the entire length of the load deprived tendons were collected. Total cellular RNA was extracted from the 20 tendons from each group by the acid guanidine thiocyanate-phenol-chloroform procedure (totally RNA kit, Ambion Inc., Austin, TX) and pooled. The RNA samples were loaded (8 jig/lane) onto a 1.2% agarose gel containing 0.66 M formaldehyde and MOPS, subjected to electrophoresis, and transferred to a nylon membrane (Pierce Corp. Rockford. IL) for 1 h in TAE at 300 mA. Following transfer, the membrane was air-dried and UV cross-linked at 10 J/cmz. The RNA blots were hybridized with a DNA probe for rat MMP-l mRNA generated in our lab and a human G3PDH cDNA control probe (Clontech Laboratories, Inc. Palo Alto, CA). The probes were labeled with biotin using the North2South Direct HRP Labeling and Detection Kit (Pierce Corp, Rockford, IL). The RNA blots were hybridized with labeled probes (10 ng/ml hybridization solution) at 55 °C for 1 h and 158 then washed. The membrane was then incubated with a chemiluminescent working solution for 5 min and exposed to films for 5-10 min. The films were scanned with a laser film digitizer (Lumiscan 75, Lumisys, Inc., Sunnyvale, CA), and MMP-1 mRNA expression was quantified by optical density measurements using Scion Image (Scion Corporation, Frederick, MD) as a ratio of G3PDH expression. The effect of load magnitude was evaluated using a Kruskal-Wallis test, followed by a Mann-Whitney U post hoc test, and the correlation between tensile load and mRNA expression of MMP-1 was examined using a polynomial regression analysis (f = I/a + bx). Significance was set at p<0.05. Western blot analysis: To verify that MMP-l mRNA expression coincided with the expression of the MMP-l protein, rat tail tendons (freshly harvested [n = 20] or load- deprived for 24 h [n = 20] as noted above) were processed for Western immunoblot staining. Total protein was extracted from each group of pooled tendons by homogenizing the tendons and lysing the cells overnight with a buffer containing 25 mM HEPES, 0.5 mM Na3V04, 50 mM sodium fluoride, 5 mM sodium pyrophosphate, 10 mM sodium B-glycerophosphate, 1 mM EGTA, 1 mM EDTA, 1 mM DTT, 1% Triton, 0.1 mM PMSF, 1 pg aprotinin, 1 uM microcystin, 1 rig/ml pepstatin, and 1 pg/ml leupeptin. The lysates were centrifuged (104 RPM at 4 °C for 30 min), and the protein quantified (BioRad Protein Assay. BioRad Labs, Hercules, CA) and electrophoresed on a 12.5% SDS-polyacrylamide gel. The separated proteins were transferred to a nitrocellulose membrane and analyzed for pro-MMP-l protein and MMP-1 protein using a mouse monoclonal antibody (Oncogene, Boston, MA) and chemiluminescence detection (New 159 England BioLabs, Beverly, MA). Human MMP-l protein (Sigma, St. Louis, MO) was used as the positive control. Actin staining/confocal laser microscopy: To verify in situ disruption of the actin cytoskeleton by cytochalasin D, additional rat tail tendons were prepared for actin staining and confocal laser microscopy. Ten millimeter segments of control (n = 5) and experimental (n = 5 treated for 1 h with 10 11M cytochalasin D) rat tail tendons were fixed in 10% phosphate buffered formalin and stained with rhodamine phalloidin (5 units/ml) (Molecular Probes, Eugene, Oregon). The tendons were wet mounted on a glass slide for viewing with a Zeiss Pascal Laser Scanning Confocal microscope (Carl Zeiss, Thornwood. NY). Observations were made using a 40x oil immersion objective with a coverslip. Fluorescent images were obtained using a HeNe 543 nm laser with a long pass 560 nm emission filter. After localizing a representative cell, a 2x zoom was used to obtain the image. Overlay images were achieved by multiple confocal 0.6 um thick '2- sectioning' through the depth of the cell and then combining the images into a single overlay image. Drugs and Chemicals: Except as noted, all drugs and chemicals (Dulbecco‘s modified Eagle medium (DMEM), fetal bovine serum (FBS), ascorbate, gentamicin, and penicillin/streptomycin-fungizone solution) were obtained from Gibco, Grand Island, New York. RESULTS The yield of total RNA extracted from each group was 0.2 rig/mg wet weight and was not significantly different among groups. 160 Freshly harvested rat tail tendons (Group 1) demonstrated no appreciable MMP-l mRNA expression on Northern blots. Tendons in the load-deprived group (Group 2) cultured for 24 h demonstrated substantial mRNA expression of MMP-1 (Figure 2). When tendons were subjected to static tensile loading during the same 24 h culture period, MMP-l mRNA expression decreased significantly with increasing load (p < 0.05) (Figure 2 and Table 1). Control 0MPa 0.16MPa 0.77MPa 1.38MPa 2.6MPa Figure A.2 Representative Northern blot gel illustrating the relative expression of MMP-1 mRNA as a function of applied stress for 24 h. G3PDH was used as an internal control. Experiments were performed three times and a representative result is shown. Table A.1 The effect of static stress on MMP-l expression STRESS MMP-l/G3PDH Fresh tendonsa Obcdef 0.00 MPa for 24 hb 0.670 : 0.266adef 0.16 MPa for 24 h“ 0.680 :1: 0.038aef 0.77 MPa for 24 hd 0.333 : 0.153ab 1.38 MPa for 24 he 0.116 t 0.050abc 2.60 MPa for 24 hf 0.077 e 0.017abc Superscripts a-e indicate Si gnificantly different corresponding data pairs (p < 0.05). 161 A strong (r2 = 0.78) and significant (p < 0.001) inverse correlation was found between the level of static tensile load and the expression of MMP-1 mRNA (Figure 3). However, MMP-l mRNA expression was not completely abolished at the highest stress (2.6 MPa) examined. Western imrnunoblot staining revealed no appreciable pro-MMP-l or MMP-l protein expression in the freshly harvested rat tail tendons. However, following 24 h of load deprivation, substantial pro-MMP-l and MMP-1 protein expression was detected in the tendons (Figure 4). Treatment with cytochalasin D did not influence MMP-l mRNA expression in the load deprived tendons (Group 2) (p > 0.05). However, cytochalasin D treatment did Significantly (p< 0.05) increase MMP-l mRNA expression in tendons exposed to a tensile loading stress of 2.60 MPa (Figure 5). Actin staining and confocal laser microscopic examination demonstrated the presence of actin stress filaments within the cells of the time zero control tendons (Figure 6A). However, in those tendons incubated for 1 h in 10 M cytochalasin D, the actin appeared globular, and no evidence of organized actin stress filaments was found (Figure 6B). 162 1.0 o #:078 o 0.8 - . p<0.0001 1175' I I 0.6 - D O. 8 E 0.4 - O. E E 0.2 . 0.0 I I I I I I 0.0 0.5 1.0 1.5 2.0 2.5 3.0 Stress (MPa) Figure A.3 Composite polynomial regression of graph of the three experimental replicates plotting MMP-l mRNA expression (normalized as a ratio of MMP-1 to G3PDH) against static stress. There was a strong (r2 = 0.78) and significant (p < 0.0001) inverse correlation between MMP-l mRNA expression and Static stress. 1 2 3 Pro-MMP1 m C 57kDa MMP1 a“: w 48kDa 1) fresh control tendon 2) 24 hr load deprived tendon 3) positive control Figure A.4 Western blot analysis illustrating the absence of pro-MMP-l and MMP-1 protein expression in freshly harvested rat tail tendons. There was a significant up-regulation of pro-MMP-l and MMP-1 protein expression in the tendon cells after 24 h of in vitro load deprivation. 163 MMP-1 G3PDH ' Figure A.5 Fresh Control 24 hr 0MPa Static Tensile Stress 24 hr 2.6MPa Static Tensile Stress 24 hr 2.6MPa Static Tensile Stress in 10uM cytochalasin Representative Northern blot gel illustrating the relative expression of MMP-1 mRNA in fresh control tendons (lane 1): 24 h load deprived at 0 MPa (lane 2); 24 h at 2.60 MPa static tensile Stress (lane 3): 24 h at 2.60 MPa static tensile Stress in 10 uM cytochalasin D (lane 4). G3PDH was used as an internal control. Experiments were performed three times and a representative result is Shown. Figure A.6 Confocal overlay images of rat tail tendon cells stained with rhodamine phalloidin to label actin filaments. (A) Fresh control tendon: note presence of actin stress fibers (arrows) in cytoskeleton. (B) Tendon treated with 10 uM cytochalasin D for 1 h; note the absence of organized actin stress fibers. (confocal 40x oil immersion: 2x zoom). 164 DISCUSSION The strain-induced signaling of cells through mechanotransduction pathways is thought to play a significant role in maintaining the normal homeostasis of connective tissues [1,8,31,34,35]. While various stress levels have been shown to induce anabolic responses in bone [12,21,22,29,30], ligament [9,20,23], and tendon cells [6,7], stress deprivation has been associated with tissue catabolism [1,3,10,16,17,30]. These catabolic changes have been attributed to degradation of the extracellular matrix by matrix metalloproteinases (MMPS) [3,16,27]. The results of the current study demonstrate that, when compared to freshly harvested control tendons, ex vivo load deprivation results in an immediate (within 24 h) increase in MMP-l mRNA expression and MMP-1 protein expression in the tendon cells. While in vivo studies have reported similar findings in immobilized ligaments and tendons [16,27], the precise mechanism by which load deprivation triggers MMP-l gene expression in these tissues is unclear. A proposed mechanism of mechanotransduction theorizes that extracellular matrix strain imparts a strain to cells through their integrin based cell-matrix connections [8,31,34,35]. The resulting cellular deformation creates changes in tension within the cytoskeleton, which can be sensed by the cell nucleus through a mechanosensory tensegrity system and results in specific cellular responses [8]. While most studies have focused on the effect of increased (over normal resting levels) strain on the mechanotransduction signaling responses of tendon cells in monolayer [6,7], the impact of total load deprivation on this process has not been examined in whole tendons ex vivo. A recent study suggested that cells are capable of establishing a constant level of tension between themselves and their extracellular matrix through a cytoskeletal tensegrity 165 system [13]. This tensional homeostasis is thought play an important role in the mechanotransduction response of the cell [13]. Removal of tendons from their normal mechanical environment could significantly alter this homeostatic tension within the cytoskeletal tensegrity system and be responsible for up—regulation of MMP-13 mRNA expression seen following 24 h of load deprivation. An in vitro study has Shown that MMP-l expression in fibroblasts is Significantly higher in free (contracting) gels than in anchored gels [25]. This suggests that the mechanical forces that are generated by the cell between itself and its surrounding matrix may, in part, regulate MMP-l expression. Sustained loss of normal cell-matrix tension, as could occur when tendons have been removed from their origins and insertions and placed in load deprived environment for 24 h, could have played a role in triggering the MMP-l expression seen in this study. Changes in cell shape, Specifically the loss of actin stress fiber organization, has been strongly correlated with collagenase gene expression [2,33]. It has been Shown that procollagenase expression in rabbit synovial fibroblasts was only induced by treatments that modified cellular actin [2,33]. This would suggest that a change in the state of actin assembly may influence collagenase expression. In the current study, treatment with cytochalasin D, a fungal product which depolymerizes actin filaments, completely abrogated the load induced inhibition of MMP-1 mRNA expression. This further supports the role of a cytoskeletally based mechanosensory tensegrity system in the control of MMP-1 mRNA expression in tendon cells. The application of a tensile load to the tendons in culture resulted in a decrease in MMP-l mRNA expression with increasing load. While MMP-l mRNA expression ex 166 vivo was decreased by increasing load, this inhibitory effect was non-linear and was not completely eliminated at the loads examined. A plausible explanation for this observation is the non-linear stress—strain behavior exhibited by rat tail tendons [18]. It is likely that with increasing load an increasing number of cells undergo nuclear (and presumably cytoskeletal) deformation [5]. Thus, the increase in the number of cells receiving a mechanotransduction stimulus with increasing load may reflect the increased inhibition of MMP-1 mRNA expression. The inability to completely eliminate MMP-l mRNA expression at the loads sampled could be explained by the fact that the loads examined in this study were all within the reported “toe” region (<3% strain) of the stress-strain curve for rat tail tendon fascicles [18]. A recent study has shown that collagen crimp within the center of the fascicles might not be completely eliminated until 2.8% strain [18]. This could mean that, in the current Study, cells within the center of the fascicle were not exposed to stresses sufficient to activate mechanotranduction responses that could affect (inhibit) MMP— 1 mRNA expression. Finally, the exact influence of an ex vivo environment on the expression of MMP- 1 mRNA in rat tail tendon cells is unknown. While the experimental design of this study would suggest that under similar ex vivo conditions static tensile loading does inhibit the expression of MMP-1 mRNA when compared to load-deprived tendons, the incomplete inhibition of this expression with what would appear to be physiologic loads may also be related to the ex vivo environment. A recent review article suggests that the absence of innervation and vascularization in explant culture systems may also contribute to the regulation of specific gene expression in connective tissues [19]. Thus, the interpretation of mechanotransduction-induced results from such culture systems must be tempered 167 with the possibility that the gene response seen ex vivo may not be reflective of that which occurs in vivo [19]. However, the effect of rigorously controlled loading and load deprivation conditions on MMP-l mRNA expression in tendon cells ex vivo seen in the current study are similar to those reported for in vivo immobilized ligaments and tendons [16,27] and in vitro cyclically loaded ligaments [27] and fibrochondrocytes [1]. A comparison of these results would suggest that the mechanostimulation of tendon cells (whether in vivo or ex vivo) does play a significant role in the regulation of MMP-1 mRNA expression. The results obtained from this ex vivo test system demonstrate that MMP-l mRNA expression in tendon cells can be affected by static tensile load, presumably through a cytoskeletally based mechanotransduction pathway. MMP-l was chosen as the gene of interest based on a previous in vitro study that documented an increase in MMP-l mRNA expression and not MMP-13 mRNA following immobilization [27]. While this study only examined the effects of various levels of static tensile load on this Signaling system, cyclic loading (which reflects a more relevant in vivo loading environment) and the resulting fluid flow have also been Shown to play a key role in the mechanotransduction response of connective tissues [4,14,23,24]. This is especially true in bone where fluid flow is believed to be an important physical Signal that influences bone cell metabolism and bone adaptation to mechanical loading [14,24,30]. The effect of cyclic load and fluid flow on the cytoskeletal components of the mechanosensory tensegrity system of tendon cells must also be investigated to completely understand the interaction of mechanical loading and tendon physiology. As in bone [21], the effect of 168 loading history on cytoskeletal adaptations and matrix interactions could provide a key to understanding the effect of various mechanical loading regimes on tendon health. 169 REFERENCES [l] Agarwal S, Long P, Gassner R, Piesco NP, Buckley MJ. Cyclic tensile strain suppresses catabolic effects of interleukin-1 beta in fibrochondrocytes from the temporomandibular joint. Arthritis Rheum 2001 ;4:608-17. [2] Aggeler J, Frisch SM, Werb Z. Changes in cell shape correlate with collagenase gene expression in rabbit synovial fibroblasts. J Cell Biol 1984;98:1662-71. [3] Amid D, Woo SL-Y, Harwood FL, Akeson W. The effect of immobilization on collagen turnover in connective tissue. A biochemical-biomechanical correlation. Acta Orthop Scand 1982;53:325-32. [4] Archambault J M, Elfervig-Wall MK, Tsuzaki M, Herzog W, Banes AJ. Rabbit tendon cells produce MMP-3 in response to fluid flow without Significant calcium transients. J Biomech 2002;35:303-9. [5] Arnoczky SP, Lavagnino M, Whallon JH, Hoonjan A. In situ cell nucleus deformation in tendons under tensile load; a morphological analysis using confocal laser microscopy. J Orthop Res 2002;20:29-35. [6] Arnoczky SP, Tian T, Lavagnino M, Gardner K, Schuler P, Morse P. Activation of stress-activated protein kinases (SAPK) in tendon cells following cyclic strain: the effects of strain frequency, strain magnitude, and cytosolic calcium. J Orthop Res 2002;20:947-52. [7] Banes AJ, Tsuzaki M, Hu P, Brigman B, Brown T, Almekinders L, et al. PDGF-BB, IGF-I, and mechanical load Stimulate DNA synthesis in avian tendon fibroblasts in vitro. JBiomech 1995:28:150.5-1513. [8] Banes AJ, Tsuzaki M, Yamamoto J, Fischer T, Brigman B, Brown T, et al. Mechanoreception at the cellular level the detection, interpretation, and diversity of responses to mechanical Signals. Biochem Cell Biol 1995;73:349-65. [9] Bhargava MM, Hannafin J A. Effect of cyclic strain on integrin expression by ligament fibroblasts. Ann Biomed Eng 1997;25:S77. [10] Bikle DD, Halloran BP. The response of bone to unloading. J Bone Miner Metab 1999;17:2334. [11] Boorman RS, Shrive NG, Frank CB. Irnmobihzation increases the vulnerability of rabbit medial collateral ligament autografts to creep. J Orthop Res 1998;16:682-9. [12] Brighton CT, Strafford B, Gross SB, Leatherwood DF. Williams JL, Pollack SR. The proliferative and synthetic response ofisolated calvarial bone cells of rats to cyclic biaxial mechanical strain. J Bone Joint Surg l99l;73A:320-3l. 170 [13] Brown RA, Prajapati R, McGrouther DA, Yannus IV, Eastwood M. Tensional homeostasis in dermal fibroblasts: mechanical responses to loading in three-dimensional substrates. J Cell Physiol 1998;175:323-32. [14] Burger EH, Kelin-Nulend J. Mechanotransduction in bone: role of the lacuno— canalicular network. FASEB J 1999;13:3101-112. [15] Burridge K, Fath K, Kelly T, Nuckolls G, Turber C. Focal adhesions: transmembrane junction between the extracellular matrix and the cytoskeleton. Ann Rev Cell Biol 1988:4z487. [16] Goomer RS, Basava D, Maris T, Kobayashi K, Harwood F, Amiel D. Effect of stress deprivation on MMP-l gene expression and regulation of MMP-1 promoter in medial collateral and anterior cruciate ligaments (MCL, ACL) and patellar tendon. Trans Orthop Res Soc 1999;24:45. [l7] Hannafin J A, Arnoczky SP, Hoonjan A, Torzilli P. Effect of stress deprivation and cyclic tensile loading on the material properties of canine flexor digitorum profundus tendons: an in vitro study. J Orthop Res 1995;13:907-14. [18] Hanson KA, Weiss J A, Barton J K. Recruitment of tendon crimp with applied tensile strain. J Biomech Eng 2002;124:72-7. [l9] Hart DA, Natsu-ume T, Sciore P, Tasevski V, Frank CB, Shrive NG. Mechanobiology: Similarities and differences between in vivo and in vitro analysis at the functional and molecular levels. Recent Res Dev Biophys Biochem 2002;2: 153-77. [20] Hsieh AH, Tsai CM-H, Ma Q-J, Lin T, Banes AJ, Villarreal FJ, et al. Time- dependent increases in type 111 collagen gene expres- Sion in medial collateral ligament fibroblasts under cyclic strain. J Orthop Res 2000; 18220-7. [21] Hsieh YF, Turner CH. Effects of loading frequency on mechanically induced bone formation. J Bone Min Res 2001;16:918-24. [22] Huiskes R, Ruimerman R, van Lenthe GH, Janssen JD. Effects of mechanical forces on maintenance and adaptation of form in trabecular bone. Nature 2000;405:704-6. [23] Hung CT, Allen FD, Pollack SR, Attia ET, Hannafin JA, Torzilli PA. Intracellular calcium response of ACL and MCL ligament fibroblasts to fluid-induced shear stress. Cell Signal l997:9: 587-94. [24] Hung CT, Pollack SR, Reilly TM, Brighton CT. Real-time calcium response of cultured bone cells to fluid flow. Clin Orthop 1995;313:25&69. 171 [25] Lambert CA, Soudant EP, Nusgens BV, Lapiere CM. Pretranslational regulation of extracellular matrix macromolecules and collagenase expression in fibroblasts by mechanical forces. Lab Invest 1992;66:44451. [26] Loitz BJ, Zemicke RF, Vailas AC, Kody MH, Meals RA. Effects of Short-term immobilization versus continuous passive motion on the biomechanical and biochemical properties of the rabbit tendon. Clin Orthop 1989;244:265-71. [27] Majima T, Matchuk LL, Shrive NG, Frank CB, Hart DA. In vitro cyclic tensile loading of an immobilized and mobilized ligament autograft selectively inhibits mRN A levels for collagenase (MMP-l). J Orthop Sci 2000;51503-10. [28] Majima T, Yasuda K, Yamamoto N. Deterioration of mechanical properties of the autograft in controlled stress-Shielded augmentation procedures. An experimental study with rabbit patellar tendon. Am J Sports Med 1994;22:821-9. [29] Neidlinger-Wilke C, Wilke H-J, Claes L. Cyclic stretching of human osteoblasts affects proliferation and metabolism: a new experimental method and its application. J Orthop Res 1994; 12170-8. [30] Rubin C, Xu G, J udex S. The anabolic activity of bone tissue, suppressed by disuse, is normalized by brief exposure to extremely low-magnitude mechanical stimuli. FASEB J 2001;] 5:2225-9. [31] Sachs F. Mechanical transduction in biological systems. Crit Rev Biomed Eng 1988;16:141-69. [32] Tremble P, Damsky CH, Werb Z. Components of the nuclear cascade that regulate collagenase gene expression in response to integrin-derived Signals. J Cell Biol 1995; 129: 1707-20. [33] Unemori EN, Werb Z. Reorganization of polymerized actin: A possible trigger for induction of procollagenase in fibroblasts cultured in and on collagen gels. J Cell Biol 1986;103:1021-31. [34] Wang N, Butler JP, Ingber DE. Mechanotransduction across the cell surface and through the cytoskeleton. Science 1993;260:1124-7. [35] Watson PA. Function follows form: generation of intracellular Signals by cell deformation. FASEB J 1991 ;5:2013-9. 172 APPENDIX B 2 2 4. 4 6 6 8 8 2 2 4 4 6 6 8 8 2 2 4. 4 6 o '2 a :4 u 06 n :8 o O .2 o .4 I .6 o .8 o o o .2 o .4 n .6 .525 .545 .565 .585 .5 6 .626 .646 .666 .686 .6 7 .727 .747 .767 . 5 . 5 . 5 . 5 . 5 . 6 . 6 . 6 . 6 . 6 . 7 . 7 . 7 . 7 I5 I I5 I I5 I ’5 I I6 I I6 I I6 I '6 I I6 I I7 I I7 I I7 I I7 I I5 5 I5 5 15 5 I5 5 I5 5 IS 5 I5 5 I5 5 I5 5 IS 5 15 5 I5 5 I5 5 I 111. 1011 (011 1011 I011 I011 I011 I011 Inualfi.l_ 1011 I011 1011 I011 I01 00000000000000000000000000000000000000000000000000000 34567890123456789012345678901234567890123456789012345 00000001111111111222222222233333333334444444444555555 1111111111111111111111111111111111111llllllllllllllll 4 4 6 6 8 8 2 2 4 4 6 6 8 8 2 2 4 4 6 6 8 8 '4 n .6 n .8 a o a :2 n 54 o .6 I .8 n a o .2 o .4 n .6 a .8 a o 2 .262 .282 .2 3 .323 .343 .363 .383 .3 4 .424 .444. .464 .484 .4 5 2 o 2 o 2 n 3 n 3 o 3 o 3 a 3 n 4 o 4 a 4 u 4 n 4 o I [2 I I2 I I3 I I3 l I3 I I3 I I3 I I4 I I4 I I4 I I4 I I4 I I5 I 5 IS 5 15 5 15 5 I5 5 I5 5 15 5 I5 5 15 5 IS 5 :5 5 IS 5 I5 5 IS 5 011 1011 I011 I011 I011 I011 I011 IOll I011 I011 1011 lOll 1011 I0 00000000000000000000000000000000000000000000000000000 01234567890123456789012345678901234567890123456789012 55555555556666666666777777777788888888889999999999000 111 m %u 1.“ rm 2 2 4. 4 6 6 8 8 2 2 4 4 6 6 8 8 2 2 mm . .2 . .4 . .6 . .8 . . .2 . .4 . .6 . .8 . . .2 . O .020 .040 .060 .080 .0 1 .121 .141 .161 .181 .1 2 .222 .24 dew. . O . 0 . O . 0 . 0 . l . l . 1 . l . .1. . 2 . 2 . dr 0 I I0 I I0 I I0 I I0 I I1 I I1 I I1 I I1 I I1 I I2 I 12 I I2 0t 5 I5 5 I5 5 I5 5 I5 5 I5 5 l5 5 I5 5 I5 5 I5 5 I5 5 I5 5 IS MS I011 1011 I011 I011 1011 I011 I011 I011 I011 I011 (011 I011 l% 0000000000000000000000000OOOOOOOOOOOOOOOOOOOOOOOO azg IIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIII l “.10.. lll1111111222222222233333333334444444444 e Gomad l m e o .tHN B S. * 173 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .15, ., 10. .05, .1, .15, ., 10.2 .05, .l, .15, ., 10.4 .05, .l, .15, 10. 10. 10. 10.2 10.2 10.2 10.4 10.4 10.4 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, ., 10.6 .05, 10.6 .1, 10.6 .15, 10.6 .,10.8 .05, 10.8 .1, 10.8 .15,10.8 ., 11. .05,11. .1,11. .15,11. ., 11.2 .05, 11.2 .1, 11.2 .15,11.2 ., 11.4 .05, 11.4 .1,11.4 .15,11.4 ., 11.6 .05, 11.6 .1, 11.6 .15, 11.6 ., 11.8 .05, 11.8 .1,11.8 .15,11.8 ., 12. .05,12. .1, 12. .15,12. ., 12.2 .05, 12.2 .1, 12.2 .15, 12.2 ., 12.4 .05, 12.4 .1, 12.4 .15, 12.4 ., 12.6 .05, 12.6 .1, 12.6 .15, 12.6 ., 12.8 .05,12.8 .1,12.8 .15,12.8 ., 13. .05, 13. .1,13. .15, 13. ., 13.2 .05, 13.2 .1, 13.2 .15, 13.2 ., 13.4 174 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .05, 13.4 .1, 13.4 .15, 13.4 ., 13.6 .05, 13.6 .1, 13.6 .15, 13.6 ., 13.8 .05, 13.8 .1, 13.8 .15, 13.8 ., 14. .05, 14. .1, 14. .15, 14. ., 14.2 .05, 14.2 .1, 14.2 .15, 14.2 ., 14.4 .05, 14.4 .1, 14.4 .15, 14.4 ., 14.6 .05, 14.6 .1, 14.6 .15, 14.6 ., 14.8 .05, 14.8 .1, 14.8 .15, 14.8 ., 15. .05, 15. .1, 15. .15, 15. ., 15.2 .05, 15.2 .1, 15.2 .15, 15.2 ., 15.4 .05, 15.4 .1, 15.4 .15, 15.4 ., 15.6 .05, 15.6 .1, 15.6 .15, 15.6 ., 15.8 .05, 15.8 .1, 15.8 .15, 15.8 ., 16. .05, 16. .1, 16. .15, 16. ., 16.2 .05, 16.2 327, 0.1, 16.2 384, 0.15, 19. 328, 0.15, 16.2 385, 0., 19.2 329, 0., 16.4 386, 0.05, 19.2 330, 0.05, 16.4 387, 0.1, 19.2 331, 0.1, 16.4 388, 0.15, 19.2 332, 0.15, 16.4 389, 0., 19.4 333, 0., 16.6 390, 0.05, 19.4 334, 0.05, 16.6 391, 0.1, 19.4 335, 0.1, 16.6 392, 0.15, 19.4 336, 0.15, 16.6 393, 0., 19.6 337, 0., 16.8 394, 0.05, 19.6 338, 0.05, 16.8 395, 0.1, 19.6 339, 0.1, 16.8 396, 0.15, 19.6 340, 0.15, 16.8 397, 0., 19.8 341, 0., 17. 398, 0.05, 19.8 342, 0.05, 17. 399, 0.1, 19.8 343, 0.1, 17. 400, 0.15, 19.8 344, 0.15, 17. 401, 0., 20. 345, 0., 17.2 402, 0.05, 20. 346, 0.05, 17.2 403, 0.1, 20. 347, 0.1, 17.2 404, 0.15, 20. 348, 0.15, 17.2 *Element, type=CAX4P 349, 0., 17.4 1, 1, 2, 6, 350, 0.05, 17.4 2, 2, 3, 7, 351, 0.1, 17.4 3, 3, 4, 8, 352, 0.15, 17.4 4, 5, 6, 10, 353, 0., 17.6 5, 6, 7, 11, 354, 0.05, 17.6 6, 7, 8, 12, 355, 0.1, 17.6 7, 9, 10, 14, 356, 0.15, 17.6 8, 10, 11, 15, 357, 0., 17.8 9, 11, 12, 16, 358, 0.05, 17.8 10, 13, 14, 18, 359, 0.1, 17.8 11, 14, 15, 19, 360, 0.15, 17.8 12, 15, 16, 20, 361, 0., 18. 13, 17, 18, 22, 362, 0.05, 18. 14, 18, 19, 23, 363, 0.1, 18. 15, 19, 20, 24, 364, 0.15, 18. 16, 21, 22, 26, 365, 0., 18.2 17, 22, 23, 27, 366, 0.05, 18.2 18, 23, 24, 28, 367, 0.1, 18.2 19, 25, 26, 30, 368, 0.15, 18.2 20, 26, 27, 31, 369, 0., 18.4 21, 27, 28, 32, 370, 0.05, 18.4 22, 29, 30, 34, 371, 0.1, 18.4 23, 30, 31, 35, 372, 0.15, 18.4 24, 31, 32, 36, 373, 0., 18.6 25, 33, 34, 38, 374, 0.05, 18.6 26, 34, 35, 39, 375, 0.1, 18.6 27, 35, 36, 40, 376, 0.15, 18.6 28, 37, 38, 42, 377, 0., 18.8 29, 38, 39, 43, 378, 0.05, 18.8 30, 39, 40, 44, 379, 0.1, 18.8 31, 41, 42, 46, 380, 0.15, 18.8 32, 42, 43, 47, 381, 0., 19. 33, 43, 44, 48, 382, 0.05, 19. 34, 45, 46, 50, 383, 0.1, 19. 35, 46, 47, 51, 175 \lmU'l \O 10 13 14 15 17 18 19 21 22 23 25 26 27 29 30 31 33 34 35 37 38 39 41 42 43 45 46 47 49 50 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 47, 49, 50, 51, 53, 54, 55, 57, 58, 59, 61, 62, 63, 65, 66, 67, 69, 70, 71, 73, 74, 75, 77, 78, 79, 81, 82, 83, 85, 86, 87, 89, 9o, 91, 93, 94, 95, 97, 98, 99, 101, 102, 103, 105, 106, 107, 109, 110, 111, 113, 114, 115, 117, 118, 119, 121, 122, 48, 50, 51, 52, 54, 55, 56, 58, 59, 60, 62, 63, 64, 66, 67, 68, 70, 71, 72, 74, 75, 76, 78, 79, 80, 82, 83, 84, 86, 87, 88, 90, 91, 92, 94, 95, 96, 98, 99, 100, 102, 103, 104, 106, 107, 108, 110, 111, 112, 114, 115, 116, 118, 119, 120, 122, 123, 52, 54, 55, 56, 58, 59, 60, 62, 63, 64, 66, 67, 68, 7o, 71, 72, 74, 75, 76, 78, 79, 80, 82, 83, 84, 86, 87, 88, 9o, 91, 92, 94, 95, 96, 98, 99, 100, 102, 103, 104, 106, 107, 108, 110, 111, 112, 114, 115, 116, 118, 119, 120, 122, 123, 124, 126, 127, 51 53 54 55 57 58 59 61 62 63 65 66 67 69 70 71 73 74 75 77 78 79 81 82 83 85 86 87 89 9O 91 93 94 95 97 98 99 101 102 103 105 106 107 109 110 111 113 114 115 117 118 119 121 122 123 125 126 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 123, 125, 126, 127, 129, 130, 131, 133, 134, 135, 137, 138, 139, 141, 142, 143, 145, 146, 147, 149, 150, 151, 153, 154, 155, 157, 158, 159, 161, 162, 163, 165, 166, 167, 169, 170, 171, 173, 174, 175, 177, 178, 179, 181, 182, 183, 185, 186, 187, 189, 190, 191, 193, 194, 195, 197, 198, 124, 126, 127, 128, 130, 131, 132, 134, 135, 136, 138, 139, 140, 142, 143, 144, 146, 147, 148, 150, 151, 152, 154, 155, 156, 158, 159, 160, 162, 163, 164, 166, 167, 168, 170, 171, 172, 174, 175, 176, 178, 179, 180, 182, 183, 184, 186, 187, 188, 190, 191, 192, 194, 195, 196, 198, 199, 128, 130, 131, 132, 134, 135, 136, 138, 139, 140, 142, 143, 144, 146, 147, 148, 150, 151, 152, 154, 155, 156, 158, 159, 160, 162, 163, 164, 166, 167, 168, 170, 171, 172, 174, 175, 176, 178, 179, 180, 182, 183, 184, 186, 187, 188, 190, 191, 192, 194, 195, 196, 198, 199, 200, 202, 203, 127 129 130 131 133 134 135 137 138 139 141 142 143 145 146 147 149 150 151 153 154 155 157 158 159 161 162 163 165 166 167 169 170 171 173 174 175 177 178 179 181 182 183 185 186 187 189 190 191 193 194 195 197 198 199 201 202 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 199, 201, 202, 203, 205, 206, 207, 209, 210, 211, 213, 214, 215, 217, 218, 219, 221, 222, 223, 225, 226, 227, 229, 230, 231, 233, 234, 235, 237, 238, 239, 241, 242, 243, 245, 246, 247, 249, 250, 251, 253, 254, 255, 257, 258, 259, 261, 262, 263, 265, 266, 267, 269, 270, 271, 273, 274, 200, 202, 203, 204, 206, 207, 208, 210, 211, 212, 214, 215, 216, 218, 219, 220, 222, 223, 224, 226, 227, 228, 230, 231, 232, 234, 235, 236, 238, 239, 240, 242, 243, 244, 246, 247, 248, 250, 251, 252, 254, 255, 256, 258, 259, 260, 262, 263, 264, 266, 267, 268, 270, 271, 272, 274, 275, 176 204, 206, 207, 208, 210, 211, 212, 214, 215, 216, 218, 219, 220, 222, 223, 224, 226, 227, 228, 230, 231, 232, 234, 235, 236, 238, 239, 240, 242, 243, 244, 246, 247, 248, 250, 251, 252, 254, 255, 256, 258, 259, 260, 262, 263, 264, 266, 267, 268, 270, 271, 272, 274, 275, 276, 278, 279, 203 205 206 207 209 210 211 213 214 215 217 218 219 221 222 223 225 226 227 229 230 231 233 234 235 237 238 239 241 242 243 245 246 247 249 250 251 253 254 255 257 258 259 261 262 263 265 266 267 269 270 271 273 274 275 277 278 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 275, 277, 278, 279, 281, 282, 283, 285, 286, 287, 289, 290, 291, 293, 294, 295, 297, 298, 299, 301, 302, 303, 305, 306, 307, 309, 310, 311, 313, 314, 315, 317, 318, 319, 321, 322, 323, 325, 326, 327, 329, 330, 331, 333, 334, 335, 337, 338, 339, 341, 342, 343, 345, 346, 347, 349, 350, 276, 278, 279, 280, 282, 283, 284, 286, 287, 288, 290, 291, 292, 294, 295, 296, 298, 299, 300, 302, 303, 304, 306, 307, 308, 310, 311, 312, 314, 315, 316, 318, 319, 320, 322, 323, 324, 326, 327, 328, 330, 331, 332, 334, 335, 336, 338, 339, 340, 342, 343, 344, 346, 347, 348, 350, 351, 280, 282, 283, 284, 286, 287, 288, 290, 291, 292, 294, 295, 296, 298, 299, 300, 302, 303, 304, 306, 307, 308, 310, 311, 312, 314, 315, 316, 318, 319, 320, 322, 323, 324, 326, 327, 328, 330, 331, 332, 334, 335, 336, 338, 339, 340, 342, 343, 344, 346, 347, 348, 350, 351, 352, 354, 355, 279 281 282 283 285 286 287 289 290 291 293 294 295 297 298 299 301 302 303 305 306 307 309 310 311 313 314 315 317 318 319 321 322 323 325 326 327 329 330 331 333 334 335 337 338 339 341 342 343 345 346 347 349 350 351 353 354 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 351, 353, 354, 355, 357, 358, 359, 361, 362, 363, 365, 366, 367, 369, 370, 371, 373, 374, 375, 377, 378, 379, 381, 382, 383, 385, 386, 387, 389, 390, 391, 393, 394, 395, 397, 398, 399, *Element, elset=High 301, 305, 309, 313, 317, 321, 325, 329, 333, 337, 341, 345, 349, 353, 357, 361, 365, 369, 352, 356, 355 354, 358, 357 355, 359, 358 356, 360, 359 358, 362, 361 359, 363, 362 360, 364, 363 362, 366, 365 363, 367, 366 364, 368, 367 366, 370, 369 367, 371, 370 368, 372, 371 370, 374, 373 371, 375, 374 372, 376, 375 374, 378, 377 375, 379, 378 376, 380, 379 378, 382, 381 379, 383, 382 380, 384, 383 382, 386, 385 383, 387, 386 384, 388, 387 386, 390, 389 387, 391, 390 388, 392, 391 390, 394, 393 391, 395, 394 392, 396, 395 394, 398, 397 395, 399, 398 396, 400, 399 398, 402, 401 399, 403, 402 400, 404, 403 type=SPRINGA, 1, 5 5, 9 9, 13 13, 17 17, 21 21, 25 25, 29 29, 33 33, 37 37, 41 41, 45 45, 49 49, 53 53, 57 57, 61 61, 65 65, 69 69, 73 373, 377, 381, 385, 389, 393, 397, 401, 405, 409, 413, 417, 421, 425, 429, 433, 437, 441, 445, 449, 453, 457, 461, 465, 469, 473, 477, 481, 485, 489, 493, 497, 501, 505, 509, 513, 517, 521, 525, 529, 533, 537, 541, 545, 549, 553, 557, 561, 565, 569, 573, 577, 581, 585, 589, 593, 597, 73, 77, 81, 85, 89, 93, 97, 101, 105, 109, 113, 117, 121, 125, 129, 133, 137, 141, 145, 149, 153, 157, 161, 165, 169, 173, 177, 181, 185, 189, 193, 197, 201, 205, 209, 213, 217, 221, 225, 229, 233, 237, 241, 245, 249, 253, 257, 261, 265, 269, 273, 277, 281, 285, 289, 293, 297, 77 81 85 89 93 97 101 105 109 113 117 121 125 129 133 137 141 145 149 153 157 161 165 169 173 177 181 185 189 193 197 201 205 209 213 217 221 225 229 233 237 241 245 249 253 257 261 265 269 273 277 281 285 289 293 297 301 177 601, 605, 609, 613, 617, 621, 625, 629, 633, 637, 641, 645, 649, 653, 657, 661, 665, 669, 673, 677, 681, 685, 689, 693, 697, 301, 305, 309, 313, 317, 321, 325, 329, 333, 337, 341, 345, 349, 353, 357, 361, 365, 369, 373, 377, 381, 385, 389, 393, 397, *Element, elset=MedHigh 302, 306, 310, 314, 318, 322, 326, 330, 334, 338, 342, 346, 350, 354, 358, 362, 366, 370, 374, 378, 382, 386, 390, 394, 398, 402, 406, 410, 414, 418, 2, 6 6, 1 10, 14, 18, 22, 26, 30, 34, 38, 42, 46, 50, 54, 58, 62, 66, 7o, 74, 78, 82, 86, 90, 94, 98, 102, 106, 110, 114, 118, 305 309 313 317 321 325 329 333 337 341 345 349 353 357 361 365 369 373 377 381 385 389 393 397 401 type=SPRINGA, O 14 18 22 26 3O 34 38 42 46 50 54 58 62 66 7O 74 78 82 86 9O 94 98 102 106 110 114 118 122 422, 426, 430, 434, 438, 442, 446, 450, 454, 458, 462, 466, 470, 474, 478, 482, 486, 490, 494, 498, 502, 506, 510, 514, 518, 522, 526, 530, 534, 538, 542, 546, 550, 554, 558, 562, 566, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, 610, 614, 618, 622, 626, 630, 634, 638, 642, 646, 122, 126, 130, 134, 138, 142, 146, 150, 154, 158, 162, 166, 170, 174, 178, 182, 186, 190, 194, 198, 202, 206, 210, 214, 218, 222, 226, 230, 234, 238, 242, 246, 250, 254, 258, 262, 266, 270, 274, 278, 282, 286, 290, 294, 298, 302, 306, 310, 314, 318, 322, 326, 330, 334, 338, 342, 346, 126 130 134 138 142 146 150 154 158 162 166 170 174 178 182 186 190 194 198 202 206 210 214 218 222 226 230 234 238 242 246 250 254 258 262 266 270 274 278 282 286 290 294 298 302 306 310 314 318 322 326 330 334 338 342 346 350 650, 350, 354 654, 354, 358 658, 358, 362 662, 362, 366 666, 366, 370 670, 370, 374 674, 374, 378 678, 378, 382 682, 382, 386 686, 386, 390 690, 390, 394 694, 394, 398 698, 398, 402 *Element, type=SPRINGA, elset=MedLow 303, 3, 7 307, 7, 11 311, 11, 15 315, 15, 19 319, 19, 23 323, 23, 27 327, 27, 31 331, 31, 35 335, 35, 39 339, 39, 43 343, 43, 47 347, 47, 51 351, 51, 55 355, 55, 59 359, 59, 63 363, 63, 67 367, 67, 71 371, 71, 75 375, 75, 79 379, 79, 83 383, 83, 87 387, 87, 91 391, 91, 95 395, 95, 99 399, 99, 103 403, 103, 107 407, 107, 111 411, 111, 115 415, 115, 119 419, 119, 123 423, 123, 127 427, 127, 131 431, 131, 135 435, 135, 139 439, 139, 143 443, 143, 147 447, 147, 151 451, 151, 155 455, 155, 159 459, 159, 163 463, 163, 167 467, 167, 171 178 471, 475, 479, 483, 487, 491, 495, 499, 503, 507, 511, 515, 519, 523, 527, 531, 535, 539, 543, 547, 551, 555, 559, 563, 567, 571, 575, 579, 583, 587, 591, 595, 599, 603, 607, 611, 615, 619, 623, 627, 631, 635, 639, 643, 647, 651, 655, 659, 663, 667, 671, 675, 679, 683, 687, 691, 695, 171, 175, 179, 183, 187, 191, 195, 199, 203, 207, 211, 215, 219, 223, 227, 231, 235, 239, 243, 247, 251, 255, 259, 263, 267, 271, 275, 279, 283, 287, 291, 295, 299, 303, 307, 311, 315, 319, 323, 327, 331, 335, 339, 343, 347, 351, 355, 359, 363, 367, 371, 375, 379, 383, 387, 391, 395, 175 179 183 187 191 195 199 203 207 211 215 219 223 227 231 235 239 243 247 251 255 259 263 267 271 275 279 283 287 291 295 299 303 307 311 315 319 323 327 331 335 339 343 347 351 355 359 363 367 371 375 379 383 387 391 395 399 699, 399, *Element, elset=Low 304, 4, 8 308, 8, 1 312, 12, 316, 16, 320, 20, 324, 24, 328, 28, 332, 32, 336, 36, 340, 40, 344, 44, 348, 48, 352, 52, 356, 56, 360, 60, 364, 64, 368, 68, 372, 72, 376, 76, 380, 80, 384, 84, 388, 88, 392, 92, 396, 96, 400, 100, 404, 104, 408, 108, 412, 112, 416, 116, 420, 120, 424, 124, 428, 128, 432, 132, 436, 136, 440, 140, 444, 144, 448, 148, 452, 152, 456, 156, 460, 160, 464, 164, 468, 168, 472, 172, 476, 176, 480, 180, 484, 184, 488, 188, 492, 192, 496, 196, 500, 200, 504, 204, 508, 208, 512, 212, 516, 216, 403 type=SPRINGA, 2 16 2O 24 28 32 36 40 44 48 52 56 60 64 68 72 76 80 84 88 92 96 100 104 108 112 116 120 124 128 132 136 140 144 148 152 156 160 164 168 172 176 180 184 188 192 196 200 204 208 212 216 220 520, 220, 524, 224, 528, 228, 532, 232, 536, 236, 540, 240, 544, 244, 548, 248, 552, 252, 556, 256, 560, 260, 564, 264, 568, 268, 572, 272, 576, 276, 580, 280, 584, 284, 588, 288, 592, 292, 596, 296, 600, 300, 604, 304, 608, 308, 612, 312, 616, 316, 620, 320, 624, 324, 628, 328, 632, 332, 636, 336, 640, 340, 644, 344, 648, 348, 652, 352, 656, 356, 660, 360, 664, 364, 668, 368, 672, 372, 676, 376, 680, 380, 684, 384, 688, 388, 692, 392, 696, 396, 700, 400, *Spring, nonlinear 0, 0 0, 0.0044 400, 20 *Spring, nonlinear 0, 0 0, 0.004 224 228 232 236 240 244 248 252 256 260 264 268 272 276 280 284 288 292 296 300 304 308 312 316 320 324 328 332 336 340 344 348 352 356 360 364 368 372 376 380 384 388 392 396 400 404 elset=High, elset=MedHigh, 179 400, 20 *Spring, elset=MedLow, nonlinear 0, 0 0, 0.0032 400, 20 *Spring, elsetzLow, nonlinear 0,0 0, 0.0006 400, 20 *Orientation, name=Ori- 1 0., 1., 0. 2, 0. ** Region: (Section- 1:Picked), (Material Orientation:Picked) *Elset, elset=_I1, generate l, 300, 1 ** Section: Section-1 *Solid Section, elset=_I1, orientation=Ori-1, materia1=YINmatrix 1., *Nset, nset=_PickedSet24, generate 4, 404, 4 *Elset, elset=_PickedSet24, generate 3, 300, 3 *Nset, nset=_PickedSet26, generate 401, 404, 1 *Elset, elset=_PickedSet26, generate 298, 300, 1 *Nset, nset=NALL, generate 1, 404, 1 *Elset, elset=NALL, generate l, 300, l *Nset, nset=_PickedSet34, generate 1, 4, 1 *Elset, elset=_PickedSet34, generate 1, 3, 1 *Nset, nset=_halfLine, generate 201, 204, 1 *‘k ** MATERIALS ** *Material, name=YINmatrix *Elastic, type=ENGINEERING CONSTANTS 0.0457, 1., 0.07769, 0.7, 0.013441, 0.1 *Permeability, specific=9.81e-06 3.1392e-09,2. ** BOUNDARY CONDITIONS ** Name: Free Edge Type: Pore pressure *Boundary _PickedSet24, 8, 8 ** Name: Radial Type: Displacement/Rotation *Boundary _PickedSet34, 2, 2 *Boundary NALL, 3, 3 *INITIAL CONDITIONS, TYPE=RATIO NALL, 2. ** 0.0457, 1.7, 0.1, ** 1% strain at 2% strain/minute ** STEP: Step-l ** *Step, nlgeom, inc=1000 *Soils, consolidation, end=PERIOD 1., 30., , , *El Print, elset=_Il, SUMMARY=YES COORD, FLVEL *CONTROLS, PARAMETERS=FIELD, FIELD=PORE FLUID PRESSURE ,,6.329E—06 ** BOUNDARY CONDITIONS ** Name: Displacement name=Step-1, amplitude=RAMP, _PickedSet26, 2, Type: Displacement/Rotation *Boundary _PickedSet26, 2, 2, 0.2 ** ** OUTPUT REQUESTS *Restart, write, frequency=1 ** ** FIELD OUTPUT: F- Output—1 ** *Output, field *Node Output U, RF, POR *Element Output S, LE, VOIDR, SAT, FLVEL ** ** HISTORY OUTPUT: H— Output-1 ** *Output, history, variable=PRESELECT *El Print, freq=999999 *Node Print, freq=999999 *FILE FORMAT, ZERO INCREMENT *End Step B.2 Global Model for 1 % strain at 20% strain/min * * .................... ** 1% strain at 20% strain/minute ** STEP: Step—1 *Step, name=Step-1, nlgeom, amplitude=RAMP, inc=1000 *Soils, consolidation, end=PERIOD 0.1, 3., , , *CONTROLS, PARAMETERS=FIELD, FIELD=PORE FLUID PRESSURE ,,6.329E-06 ** BOUNDARY CONDITIONS ** Name: Displacement Type: Displacement/Rotation *Boundary 180 B.3 Global Model for 3% strain at 6% strain/minute ** ** 3% strain at 6% strain/minute ** STEP: Step-1 *Step, name=Step-1, nlgeom, amplitude=RAMP, inc=1000 *Soils, consolidation, end=PERIOD 1., 30., , , *CONTROLS, PARAMETERS=FIELD, FIELD=PORE FLUID PRESSURE ,,6.329E-06 ** BOUNDARY CONDITIONS ** Name: Displacement Type: Displacement/Rotation *Boundary _PickedSet26, 2, 2, 0.6 BA Global Model for 3% strain at 2% strain/minute ** 3% strain at 2% strain/minute ** STEP: Step-1 *Step, name=Step-1, nlgeom, amplitude=RAMP, inc=1000 *Soils, consolidation, end=PERIOD 1., 130., , , *CONTROLS, PARAMETERS=FIELD, FIELD=PORE FLUID PRESSURE ,,6.329E-06 ** BOUNDARY CONDITIONS ** Name: Displacement Type: Displacement/Rotation *Boundary _PickedSet26, 2, ———~—-———————a—————~a— B.5 Submodel for 1% strain at 2% strain/minute *Heading *Node 1, 0.12, 9.89 2, 0.1225, 9.89 3, 0.1225, 9.91 4, 0.12, 9.91 5, 0.13, 9.89 6, 0.1275, 9.89 7, 0.1275, 9.91 8, 0.13, 9.91 9, 0.127, 9.91 10, 0.123, 9.91 11, 0.123, 9.89 12, 0.127, 9.89 13, 0.1, 9.8 14, 0.15, 9.8 15, 0.15, 10. 16, 0.1, 10. 17, 0.1273756, 9.889221 18, 0.1270097, 9.888513 19, 0.1264553, 9.887967 20, 0.125782, 9.887626 21, 0.1250419, 9.887501 22, 0.1243278, 9.887592 23, 0.1237166, 9.887855 24, 0.123218, 9.888247 25, 0.1228318, 9.888756 26, 0.1225847, 9.889355 27, 0.1225, 9.891253 28, 0.1225, 9.892502 29, 0.1225, 9.893751 30, 0.1225, 9.895 31, 0.1225, 9.89625 32, 0.1225, 9.8975 33, 0.1225, 9.89875 34, 0.1225, 9.9 35, 0.1225, 9.90125 36, 0.1225, 9.9025 37, 0.1225, 9.90375 38, 0.1225, 9.904999 39, 0.1225, 9.906248 40, 0.1225, 9.907496 41, 0.1225, 9.908743 42, 0.122596, 9.910686 43, 0.1228836, 9.911331 44, 0.1233149, 9.911847 45, 0.1238201, 9.912204 46, 0.1245113, 9.912452 47, 0.1252149, 9.912491 48, 0.1259368, 9.912318 49, 0.1265545, 9.911958 50, 0.1270634, 9.911411 51, 0.1273919, 9.910728 52, 0.1275, 9.908745 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1275, .1275, .1275, .1275, .1275, .1275, .1275, .1275, .1275, .1275, .1275, .1275, .1275, .1275, .1201603, 9 .1206616, .1214864, .1225794, .1238598, .1252736, .1266752, .1279844, .1290676, .1297744, 9 .13, 9. .892498 .893749 .895 .89625 .897499 .89875 .9 .90125 .9025 .903751 .905005 .906258 .907513 .13, 9. .13, .13, .13, .13, .13, .13, .13, .13, .13, .13, .13, .13, .13, .129833, .129309, .1284311, .1272283, .1258984, .1244844, .1230736, .1218183, 9. .1207713,9. .1201865,9. 9. 0000000000 \OKDKOKJKOKDKOKDWWWKDKO .12, .12, .12, .12, .12, .12, .12, .12, \DKDKOKOkOKOLO WOKOkOtOkOKDKDODKOkOKD \0 .907496 .906248 .905 .90375 .9025 .90125 .9 .89875 .8975 .89625 .895 .89375 .892503 .891255 .888744 .887514 .886443 .885625 .885132 .885008 .885289 .885988 .887093 .888515 891246 koxoxoxotokoxoko 908789 9.911282 9.912537 .913637 .914476 .914919 .914973 .914614 913857 912668 911353 908752 .907501 .90625 .905 .90375 .9025 .90125 .900001 \OKDKOKOKO 1'81 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .12, .12, .12, .12, .12, .12, .12, 9 \DKOKDKDLDKD .89875 .8975 .89625 .895 .89375 .8925 .891251 .1230789,9. .1233067,9. .1236304,9. .1239892,9. .124416, .1250051 .1255653,9.888082 9. 9.888 I 889443 888936 888542 888274 888087 .1260631,9.888306 .126531, 9.888713 .1268786,9.889314 .127, 9.891258 .127, 9.892504 .127, 9.893751 .127, 9.895 .127, 9.89625 .127, 9.8975 .127, 9.89875 .127, 9.9 .127, 9.90125 .127, 9.9025 .127, 9.90375 .127, 9.905 .127, 9.906248 .127, 9.907495 .127, 9.908741 .1268968,9.910634 .1265889,9.911215 .1261494,9.911636 .1256743,9.911883 .1251192,9.911997 .1245382,9.911946 .1239607,9.911709 .1236536,9.911479 .1233289,9.911098 .1230874,9.910584 .123, 9.908737 .123, 9.907492 .123, 9.906246 .123, 9.904999 .123, 9.903749 .123, 9.9025 .123, 9.90125 .123, 9.9 .123, 9.89875 .123, 9.8975 .123, 9.89625 .123, 9.895 .123, 9.893751 .123, 9.892506 .123, 9.891265 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .105, .11, .115, .12, .125, .13, .135, .14, .145, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, .15, 9 9. 9. 9. 9 9. 9. 9. \OKDOKOkDKOKDkDKOKDKDKDkaDkD\DkaOWKDKOKOKO\DKOKOkOKDkO\D\O\O\O\O\O\O\OKO\O\OKO\DKOKOKO\O\O\O 9 .8 8 8 8 .8 8 8 8 .8 .805024 .81005 .815056 .820071 .825154 .830199 .835218 .840233 .84523 .850204 .855147 .860065 .865031 .869888 .874309 .878421 .881973 .885245 .887938 .890355 .892571 .89453 .896206 .897494 .898749 .900001 .901252 .902507 .903771 .90537 .907381 .909738 .912341 .915176 .918248 .92154 .924954 .928956 .932903 .936989 .941261 .94559 .950184 .954956 .959836 .964806 .9698 .974839 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 243, 244, 245, 246, 247, 248, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, H OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .15, .15, .15, .15, .1464286, .1428571, .1392857, .1357143, .1321429, .1285714, .125, .1214286, .1178572, .1142857, .1107143, .1071428, .1035714, 9. \ ‘ S ‘ ‘ ‘ § § § ‘ ~ ‘ ‘ ‘ § § ‘ § \ ‘ ‘ \ \ ‘ ‘ ‘ ‘ ‘ ‘ ~ ‘ ‘ ‘ ~ ‘ ~ ‘ H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H m m m m m m m m m m m m m m m m m m m m m m w m m m m m m m m m m m m m m m m ‘ 9 9. 9 9. .98994 10. 994998 .984993 .980019 .975111 .970166 .965195 .960217 .950606 .946121 .94188 .937828 .933922 .930047 .926426 .923129 .920083 .917194 .914449 .911864 .909427 .907158 .905291 .903801 .902511 .901254 .900002 .89875 .897498 .896176 .894773 .892802 .890473 .887895 .88503 .881431 .877465 .872651 .867185 .861723 .85646 182 .979858 984921 994965 10. 10. 10. 10. 10. 10. 10. 10. 10. 10. 10. 10. 283, 284, 285, 286, 288, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 340, 341, .1, 9. .1208519,9. .1216696,9. .1216717,9. .1208918,9. .1240684,9. .124189, .1263034,9. .1266969,9. .1290812, .1283246,9. .1283117,9. .1290788,9. .1208389,9. .1216691,9. .1208351,9. .1216678,9. .1208339,9. .1216671,9. .1208335, .1216668, .1208334, .1216667, .1208334, .1216667, .1208333, .1216667, .1208333,9.900001 .1216667,9.900001 .1208333, .1216667, .1208333, .1216667, .1208334, .1216667, .1208336, .1216669,9. .1208348,9. .1216678,9. .1208388,9. .1216699,9. .1208509,9. .1216731,9. .1230566,9. .1233874,9. .1221911,9. .122702, .1215272,9. .1221746,9. .1210895,9. OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO ‘ ‘ I I H H H H H H H m w m m m m m I 9 9 .851249 .846066 .84089 .835728 .825469 .815261 .810154 805083 890019 890038 909966 909966 88616 .886895 9 9 9 9 9. 9 9 9 9 9 9 9 9. 9 9 9 912914 913556 .89002 890027 909901 909927 891259 891264 892504 892506 893752 893753 .895 .895 .89625 89625 .8975 .8975 .89875 .89875 .90125 .90125 .9025 9025 .90375 .90375 .905 904999 906248 906247 907495 907494 908737 908735 886545 887211 887185 .887729 888024 888406 888994 342, 343, 344, 345, 346, 347, 348, 349, 350, 351, 352, 353, 354, 355, 356, 357, 358, 359, 360, 361, 362, 363, 364, 365, 366, 367, 368, 369, 370, 371, 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384, 385, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396, 397, 398, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1218188, .1254165,9.913175 .1256191,9.913901 .1245016,9.913165 .1245097,9.91393 .1236368,9.912906 .l234176,9.913612 .122876, 9. .1224437,9.912994 .1222797,9.911696 .121662, 9. .1218823,9.910859 .121153, 9. .1250973,9.886806 .1251706,9.886068 .1260372,9.887 .1262947,9.886303 .1268941,9.887464 .1273307,9.886866 .1275871,9.888171 .1281903,9.887741 .1280564, .1287756,9.888834 .1276392, .1270733,9.912433 .1283716,9.912064 .1276964,9.9ll732 .1288473,9.911034 .1281042,9.910854 .1291413,9.908737 .1283204,9.908719 .1291585,9.907499 .1283276,9.907492 .129164, 9. .128331, 9. .1291657,9.905003 .1283325,9.905001 .1291663,9.903751 .128333, 9. .1291666, .1283332, .l29l666,9.901249 .1283333,9.901249 .1291667, .1283333, .1291667, .1283333, .1291667,9.897499 .1283333,9.897499 .1291666, .1283332, .129166, .1283327, .1291641,9.893751 .1283313,9.893751 .1291583,9.892503 .1283283,9.892504 9.88919 912401 912106 911061 9.88906 9.91293 906252 906248 90375 9.9025 9.9025 9.9 9.9 9.89875 9.89875 9.89625 9.89625 9.895 9.895 399, 400, 401, 402, 403, 404, 405, 406, 407, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, 431, 432, 433, 434, 435, 436, 437, 438, 439, 440, 441, 442, 443, 444, 445, 446, 447, 448, 449, 450, 451, 452, 453, 454, 455, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1291407,9.891257 .1283234,9.89126 .1245945,9.909866 .1245919,9.890188 .126192, 9. .1253887,9.909861 .1238021,9.909926 .1253856,9.890137 .1261894,9.890067 .1238036,9.890131 .1245898,9.911556 .1245674,9.911187 .1245813,9.910816 .1245873,9.910376 .124534, 9. .1245284,9.888803 .12453, .1245648,9.889647 .1261965,9.908709 .125395, .1245969, .1237996,9.908705 .1261985,9.907479 .1253978,9.907467 .1245984,9.907465 .1237994,9.907475 .1261993, .125399, 9. .1245992,9.906233 .1237996,9.906237 .1261997,9.904997 .1253996,9.904993 .1245996,9.904992 .1237998,9.904994 .1261999,9.903749 .1253998,9.903748 .1245998,9.903747 .1237999,9.903748 .1262, .1253999,9.902499 .1245999,9.902498 .1237999,9.902498 909928 88844 9.889205 9.908681 9.90868 9.90624 906235 9.902499 .1262, 9.901249 .1254, 9.901249 .1246, 9.901249 .1238, 9.901249 .1262, 9.9 .1254, 9.9 .1246, 9.9 .1238, 9.9 .1262, 9.89875 .1254, 9.89875 .1246, 9.89875 .1238, 9.89875 .1262, 9.897499 .1254, 9.897499 .1246, 9.897499 183 456, 457, 458, 459, 460, 461, 462, 463, 464, 465, 466, 467, 468, 469, 470, 471, 472, 473, 474, 475, 476, 477, 478, 479, 480, 481, 482, 483, 484, 485, 486, 487, 488, 489, 490, 491, 492, 493, 494, 495, 496, 497, 498, 499, 500, 501, 502, 503, 504, 505, 506, 507, 508, 509, 510, 511, 512, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1238, .1262, .1254, .1246, .1238, 9. .1261999, .1253998,9.895001 .1245998,9.895002 .1237999,9.895002 .1261994,9.893755 .1253991, .1245993,9.893763 .1237997,9.893761 .1261982,9.892517 .1253973,9.892532 .124598, 9. .1237994, .1261953,9.891291 .1253935,9.891325 .1245958,9.891342 .1237997,9.891317 .1254858,9.911613 .1250823,9.911595 .1260817,9.910472 .1258958,9.910967 .1256716,9.911354 .1251406,9.911226 .1252392,9.910832 .1253242,9.910385 .1241713,9.911494 .1240816, .1239427,9.910886 .1238549,9.910442 .1241872,9.888486 .1238417, .1239246,9.889132 .1240677,9.888741 .1254048,9.888359 .1250043,9.888385 .1256022,9.888604 .125843, 9. .1260521,9.889493 .1252972,9.889603 .125187, 9. .1250805,9.888731 .1251679,9.855898 .1249347,9.946147 .1201744, .1151425, .1101005,9.856346 .1050535,9.856426 .1450486,9.855179 .1400837,9.855508 .1351184,9.855652 .1301433,9.855773 .12525, .1253804,9.865154 .897499 .89625 .89625 .89625 89625 9.895 \OUJLOKO 9.89376 89254 9.89253 9.91126 9.88959 888987 889132 9.8561 9.85638 9.860677 513, 514, 515, 516, 517, 518, 519, 520, 521, 522, 523, 524, 525, 526, 527, 528, 529, 530, 531, 532, 533, 534, 535, 536, 537, 538, 539, 540, 541, 542, 543, 544, 545, 546, 547, 548, 549, 550, 551, 552, 553, 554, 555, 556, 557, 558, 559, 560, 561, 562, 563, 564, 565, 566, 567, 568, 569, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1255717,9.869333 .1257468,9.873148 .1257962,9.876843 .1256901,9.879618 .1255477,9.881708 .1254307,9.883379 .1175374,9.889952 .1154984,9.889482 .1134595,9.888799 .1113164,9.887873 .1089269,9.886832 .1062072,9.885845 .1031775,9.885231 .1468403,9.885336 .1437661, .1408746,9.886351 .1383104,9.887238 .1360513,9.888116 .1340202,9.888878 .1321107,9.889471 .1176932,9.910164 .1159885,9.910698 .1141984,9.911491 .1120286,9.912323 .1095356,9.913473 .1063889,9.914026 .1031867,9.914318 .1464794,9.914962 .1430041,9.914434 .140153, 9. .1377452,9.912246 .1356355,9.911454 .1337084, .1318848, .1244525,9.915964 .1244569, .1244739,9.918481 .1244928,9.920108 .12452, .1245608,9.923951 .1246456,9.926343 .1247237,9.928861 .1247986,9.931556 .1248462,9.934562 .124881, 9. .1249095,9.942087 .1213575,9.946206 .1177838,9.946188 .114214, 9. .1106495,9.946076 .107092, 9. .1035435,9.946094 .1464283,9.945506 .142849, 9. .1392655, .1356802,9.945755 .1320961,9.945908 9.88573 913254 9.91084 9.91037 9.91704 9.921928 938158 946131 946071 945532 9.94561 570, 571, 572, 573, 574, 575, 576, 577, 578, 579, 580, 581, 582, 583, 584, 585, 586, 587, 588, 589, 590, 591, 592, 593, 594, 595, 596, 597, 598, 599, 600, 601, 602, 603, 604, 605, 606, 607, 608, 609, 610, 611, 612, 613, 614, 615, 616, 617, 618, 619, 620, 621, 622, 623, 624, 625, 626, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1285141,9.946049 .1050278,9.851227 .1050137,9.846064 .1050064,9.840889 .1050027,9.835724 .1050008, .1050003, .1050001,9.820381 .1050001,9.815293 .105, .105, .110044, 9. .1100205,9.846174 .1100092,9.841063 .110004, 9. .1100017,9.830806 .1100007,9.825674 .1100003,9.820541 .1100001,9.815409 .11, .11, .1150731,9.851378 .1150368, .1150155, .1150055,9.836206 .1150024,9.831039 .115001, .1150004,9.820691 .1150002,9.815516 .1150001,9.810341 .115, .1201007,9.851l98 .1200551,9.846235 .1200248,9.841275 .1200087,9.836271 .1200039,9.831108 .1200011,9.825985 .1200005, .1200002,9.815578 .1200001,9.810379 .12, .1251042,9.851041 .125058, .1250272, .1250118,9.836168 .1250052,9.831052 .1250015,9.825944 .1250005,9.820782 .1250002,9.815576 .1250001,9.810375 .125, .l300901,9.850926 .1300507,9.846029 .1300237,9.841093 .1300096,9.836077 .1300037,9.830986 .1300011,9.825873 9.83056 9.82547 9.810202 9.805104 851257 835938 9.810274 9.805138 9.84633 9.84129 9.825866 9.805168 9.82078 9.805185 9.84614 9.8412 9.805183 184 627, 628, 629, 630, 631, 632, 633, 634, 635, 636, 637, 638, 639, 640, 641, 642, 643, 644, 645, 646, 647, 648, 649, 650, 651, 652, 653, 654, 655, 656, 657, 658, 659, 660, 661, 662, 663, 664, 665, 666, 667, 668, 669, 670, 671, 672, 673, 674, 675, 676, 677, 678, 679, 680, 681, 682, 683, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1300005,9.820687 .1300002,9.815505 .1300001,9.810329 .13, .1350663,9.850823 .1350309,9.845955 .1350116,9.841017 .1350055,9.835914 .1350025,9.830791 .1350012,9.825657 .l350005,9.820522 .1350002,9.815388 .1350001,9.810254 .135, .1400378,9.850653 .1400195,9.845636 .1400098,9.840592 .1400047,9.835531 .1400022,9.830462 .140001, 9. .1400005,9.820328 .1400002,9.815252 .1400001, .14, .1450245, .1450125,9.845258 .1450063,9.840252 .1450031,9.835237 .1450015,9.830225 .1450007,9.825193 .1450003,9.820142 .1450001,9.815135 .1450001,9.810101 .145, .1107116,9.873552 .1101853,9.861538 .1103606,9.867079 .104999, .1208478, .1161493,9.871035 .1202417,9.885108 .1191315,9.883805 .117807, .1162192, .1139727,9.877419 .1070493, .1083724,9.878796 .1051043,9.867197 .1050857,9.861694 .1152809,9.861296 .1202895,9.860893 .1155376,9.866225 .1204571,9.865458 .1216608,9.873728 .1223483,9.877121 .1228565,9.879744 .1232394,9.881777 9.805161 9.805124 825393 9.81017 9.805085 9.85024 9.805052 9.872904 9.8696 9.882193 9.8801 9.88258 684, 685, 686, 687, 688, 689, 690, 691, 692, 693, 694, 695, 696, 697, 698, 699, 700, 701, 702, 703, 704, 705, 706, 707, 708, 709, 710, 711, 712, 713, 714, 715, 716, 717, 718, 719, 720, 721, 722, 723, 724, 725, 726, 727, 728, 729, 730, 731, 732, 733, 734, 735, 736, 737, 738, 739, 740, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1235388,9.883355 .ll77046,9.875055 .1190799, .1201908,9.880621 .1210629,9.882492 .1217759,9.883947 .1189644,9.886529 .1180427,9.888242 .1175222,9.885546 .1163976,9.887438 .1158578,9.884308 .1145351,9.886467 .1140171,9.88262 .1124644,9.8852l9 .1115812,9.880709 .1100044,9.883847 .1036167,9.881796 .1042433,9.877834 .1356599, .1351836,9.860476 .1352672,9.865288 .1354349,9.870108 .1302797, .1456014,9.874557 .1410072,9.875127 .1398581,9.883514 .1383183,9.880283 .1290355,9.884704 .1300215,9.883231 .1312946,9.881398 .13302, .1303119, .1302496,9.865171 .1302034,9.860549 .1401481,9.860394 .1450936,9.860091 .1402809,9.865186 .1451795,9.865044 .1405316,9.870129 .1453342,9.87 .1460573,9.878643 .146491, 9. .1421306,9.879326 .1430666,9.882719 .137173, 9 .134959, 9. .1331235,9.886932 .1315948,9.887863 .1355158,9.882025 .1334493,9.883652 .1318114,9.884992 .1305189,9.886071 .1273108,9.883775 .12786, .1285599,9.880175 .1294116,9.877542 .1028988,9.888037 9.87817 9.87584 9.87397 9.879214 9.86943 882168 .884703 885872 9.882222 741, 742, 743, 744, 745, 746, 747, 748, 749, 750, 751, 752, 753, 754, 755, 756, 757, 758, 759, 760, 761, 762, 763, 764, 765, 766, 767, 768, 769, 770, 771, 772, 773, 774, 775, 776, 777, 778, 779, 780, 781, 782, 783, 784, 785, 786, 787, 788, 789, 790, 791, 792, 793, 794, 795, 796, 797, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1057024,9.888452 .1083303,9.889072 .110756, 9. .113008, 9. .1152467,9.890943 .1175055,9.891241 .1027154, .1053734, .1079364,9.891212 .1103702,9.891693 .1127083,9.892159 .1151284,9.892315 .1175262,9.892462 .1025909,9.892895 .1051434,9.893103 .1076773,9.893194 .1101042,9.893513 .1125244,9.893699 .1150326,9.893698 .1175051,9.893741 .102519, 9. .1050144,9.894938 .1075372,9.894886 .1100169,9.894957 .1125119,9.894973 .1150068,9.894985 .1175032,9.894994 .1025028,9.896231 .1050056,9.896225 .1075071,9.896226 .1100071,9.896231 .1125055,9.896236 .1150035,9.896242 .1175018,9.896246 .102501, 9. .1050021,9.897491 .1075028,9.897491 .1100029,9.897491 .1125024,9.897494 .1150017,9.897496 .1175009,9.897498 .1025004,9.898748 .1050008,9.898746 .1075011,9.898746 .1100012,9.898746 .112501, .1150008,9.898748 .1175004,9.898748 .1025001, .1050003,9.899999 .1075004,9.899999 .1100005,9.899999 889786 890496 9.89057 9.89085 894836 897493 9.898748 9.9 .1125004, 9.9 .1150003, 9.9 .1175002, 9.9 .1025, 9.901251 .1050001, 9.90125 185 798, 799, 800, 801, 802, 803, 804, 805, 806, 807, 808, 809, 810, 811, 812, 813, 814, 815, 816, 817, 818, 819, 820, 821, 822, 823, 824, 825, 826, 827, 828, 829, 830, 831, 832, 833, 834, 835, 836, 837, 838, 839, 840, 841, 842, 843, 844, 845, 846, 847, 848, 849, 850, 851, 852, 853, 854, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1075002, .1100002, .1125002, .1150001, .1175001, .1025, .105, .1075001, .1100001, .1125001, .1150001, .1175, .1025002,9. .1050006,9. .1075008,9. .1100009,9. .1125007,9. .1150005,9. .1175003,9. .1025188, .1050073,9. .1075115,9. .1100128,9. .1125082,9. .1150043,9. .1175021,9. .1026081, .1051775,9. .1077127,9. .1101901,9. .1126029,9. .115039, .1175182,9. .1027591,9. .1054776,9. .108112, .1106027,9. .1129512,9. .115213, .1175589,9. .1029544,9. .105872, .1086677,9. .1111743,9. .1134557, .1155313,9. .1176096,9. .146859, .1438222,9. .1410762,9. .1386052,9. .1363501,9. .1342404,9. .1321494,9. .1471345,9. .1443491,9. .1417162,9. \O\O&D\D 9. 9.902504 9.902501 9 9 9 9 9 9 9. 9. 9. 9. 9 9. .90125 .90125 .90125 .90125 90125 .9025 .9025 .9025 .9025 9.9025 903763 903757 903753 903752 903751 903751 903751 .9052 905041 905033 905025 905014 905006 905003 .90706 906847 906702 906548 906378 906293 906269 909286 909056 908765 908403 908028 907703 907555 911718 911434 910966 910325 .90975 909206 908844 912149 911717 911035 910375 909803 909328 908999 909578 909268 908859 855, 856, 857, 858, 859, 860, 861, 862, 863, 864, 865, 866, 867, 868, 869, 870, 871, 872, 873, 874, 875, 876, 877, 878, 879, 880, 881, 882, 883, 884, 885, 886, 887, 888, 889, 890, 891, 892, 893, 894, 895, 896, 897, 898, 899, 900, 901, 902, 903, 904, 905, 906, 907, 908, 909, 910, 911, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1392237, .1369032, .1345779, .1323334, .1473269, .1447156, .1421779, .1396701, .1372803, .1348348, .1324085, .1474449, .1449248, .1423945, .139882, .1373917, .134916, .1324519, .1474851, .1449679, .1424535, .1399461, .1374485, .1349587, .1324756, .147494, .1449867, .1424801, .1399762, .1374763, .1349803, .1324881, .1474977, .1449947, .1424918, .1399898, .1374895, .1349909, .1324944, .1474991, .1449979, .1424967, .1399957, .1374954, .1349959, .1324974, .1474997, .1449992, .1424987, .1399982, .1374981, .1349982, .1324988, .1474999, .1449997, .1424995, .1399993, 9.908501 9.908091 9.90786 9.907627 9.907253 9.907004 9.906796 9.906679 9.906474 9.906397 9.906328 9.905268 9.905179 9.905178 9.905155 9.905119 9.905081 9.905045 9.903806 9.903826 9.903831 9.903824 9.90381 9.903793 9.903774 9.902521 9.902531 9.902535 9.902534 9.902529 9.902521 9.902513 9.901258 9.901262 9.901265 9.901264 9.901263 9.901259 9.901256 9.900002 9.900005 9.900005 9.900005 9.900005 9.900004 9.900002 9.89875 9.898751 9.898751 9.898751 9.898751 9.898751 9.89875 9.897497 9.897499 9.897499 9.897499 912, 913, 914, 915, 916, 917, 918, 919, 920, 921, 922, 923, 924, 925, 926, 927, 928, 929, 930, 931, 932, 933, 934, 935, 936, 937, 938, 939, 940, 941, 942, 943, 944, 945, 946, 947, 948, 949, 950, 951, 952, 953, 954, 955, 956, 957, 958, 959, 960, 961, 962, 963, 964, 965, 966, 967, 968, OOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOOO .1374992, .1349992, .1324995, .1474999, .1449997, .1424995, .1399993, .1374991, .1349991, .1324994, .1474661, .1449473, .142452, .1399944, .1374937, .1349947, .1324968, .1473832, .1447983, .1422724, .1398373, .1373949, .1349758, .1324874, .1472633, .1445704, .1419716, .1394886, .1371044, .1348103, .1324629, .1470879, .1442408, .141535, .139021, .1366829, .1345284, .1323652, .1110833, .1106174, .1105716, .1105101, .1104221, .1103808, .1104783, .1106029, .1149103, .1182574, .1214442, .1034042, .106967, .1185814, .1171023, .1154164, .1134585, .1212621, .121196, 186 9.897499 9.897499 9.897499 9.896242 9.896246 9.896248 9.896248 9.896248 9.896248 9.896249 9.89467 9.894751 9.89485 9.894987 9.894989 9.894991 9.894995 9.892652 9.892828 9.893066 9.893411 9.893575 9.893715 9.893732 9.890459 9.890674 9.891031 9.891451 9.89185 9.892241 9.89245 9.888027 9.888317 9.888821 9.889437 9.890028 9.890647 9.891068 9.916774 9.941801 9.937749 9.933907 9.930395 9.927011 9.923742 9.920314 9.918882 9.920525 9.921429 9.917135 9.917007 9.911798 9.91249 9.913341 9.914528 9.92361 9.926044 969, 0 970, 0.1212301, 971, 0 972, 0 973, 0.1213313, 974, 0.1178275,9. 975, 0.1176724,9. 976, 0.1176279,9. 977, 0.1176399,9. 978, O.1l76819,9. 979, 0.1177211,9. 980, 0.1177553,9. 981, 0.1142109,9. 982, 0.1140854,9. 983, 0.1140319,9. 984, O.1140439,9. 985, 0.1140949,9. 986, 0.1141435,9. 987, 0.1141831,9. 988, 0.1070629,9. 989, 0.1035266,9. 990, 0.1070143,9. 991, 0.1035052,9. 992, 0.1069824,9. 993, 0.1034856,9. 994, 0.1069362,9. 995, 0.1034629,9. 996, 0.1069171,9. 997, 0.1034426,9. 998, 0.1069607,9. 999, 0.1034595,9. 1000,0.1069961,9. 1001, 0.103464,9. 1002,0.1195546,9. 1003,0.1209963,9. 9. 1005,0.1185653,9. 1006,0.1203863,9. 1007,0.1223928,9. 1008,0.1174505,9. 1009,0.1196402,9. 1010,0.1220636,9. 9. 1012,0.1189316,9. 1013,0.1217346,9. 1014,0.1415594,9. 1015,0.1428448,9. 9. 1017,0.1428255,9. 1018,0.1427504,9. 1019,0.1426704,9. 1020,0.1422999,9. 1021,0.1460539,9. 1022, 0.127765,9. 1023,0.1310084,9. 1024,0.1343313,9. 1025,0.1376302,9. 1004, 0.12266, 1011, 0.11618, 1016,0.142837, .1211978,9.928662 9.9314 .1212665,9.934532 .1213007,9.938177 9.9421 922902 925529 928276 931114 934359 938074 942044 921573 924613 927666 930777 934119 937905 941934 941694 941738 937531 937726 933874 933891 930239 930131 926708 926417 923421 923139 920171 920097 913328 914688 915543 914233 915812 916625 915383 917114 918036 916845 918587 919628 917841 941188 937016 932974 929192 925168 921541 918103 923653 923018 921722 920015 1026,0. 1027,0. 1028,0. 1029,0. 1030,0. 1031,0. 1032,0. 1033,0. 1034,0. 1035,0. 1036,0. 1037,0. 1038,0. 1039,0. 1040,0. 1041,0. 1042,0. 1043,0. 1044,0. 1045,0. 1046,0. 1047,0. 1048,0. 1049,0. 1050,0. 1051,0. 1052,0. 1053,0. 1054,0. 1055,0. 1056,0. 1057,0. 1058,0. 1059,0. 1060,0. 1061,0. 1062,0. 1063,0. 1064,0. 1065,0. 1066,0. 1067,0. 1068,0. 1069,0. 1070,0. 1071,0. 1072,0. 1073,0. 1074,0. 1075,0. 1076,0. 1077,0. 1078,0. 1079,0. 1080,0. 1081,0. 1082,0. 1314439,9. 1329769,9. 1346569,9. 1365484,9. 1387843,9. 1461682,9. 1463128,9. l463679,9. 1464084,9. 1464244,9. 1464287,9. 1392518,9. 1356608,9. 1320736,9. 1284903,9. 1392321,9. 1356352,9. l320458,9. 1284621,9. 1392018,9. 1355977,9. l32007l,9. 1284265,9. 1391282,9. 911715 912303 913056 914021 915322 921516 925011 929011 932888 93692 941099 941419 941644 941847 941992 937354 93765 937901 938078 933404 933841 934223 934469 929751 135519,9.930481 1319293,9. 1283422,9. 1389619,9. 1352943,9. 13l7161,9. 1281977,9. 1384688,9. 1349217,9. 1314262,9. 1280086,9. 1274812,9. 1271712,9. 1268308,9. 1264964,9. 1262256,9. 1304469,9. 1298318,9. 1291327,9. 1284585,9. 1279045,9. 1333929,9. 1324131,9. 1313228,9. 1302775,9. 1293766,9. 1360908,9. 1345556,9. 1330992,9. 931128 931465 926232 927382 928174 928631 923016 924455 925477 926026 921614 919862 91831 91694 915874 920905 919176 917668 916419 915353 91951 917771 916416 915313 91439 917727 916061 914828 131775,9.913864 1305955,9. 1464295,9. 1464287,9. 913102 949978 954803 1083,0. 1084,0. 1085,0. 1086,0. 1087,0. 1088,0. 1089,0. 1090,0. 1091,0. 1092,0. 1093,0. 1094,0. 1095,0. 1096,0. 1097,0. 1098,0. 1099,0. 1100,0. 1101,0. 1102,0. 1103,0. 1104,0. 1105,0. 1106,0. 1107,0. 1108,0. 1109,0. 1110,0. 1111,0. 1112,0. 1113,0. 1114,0. 1115,0. 1116,0. 1117,0. 1118,0. 1119,0. 1120,0. 1121,0. 1122,0. 1123,0. 1124,0. 1125,0. 1126,0. 1127,0. 1128,0. 1129,0. 1130,0. 1131,0. 1132,0. 1133,0. 1134,0. 1135,0. 1136,0. 1137,0. 1138,0. 1139,0. 1464286,9. 1464286,9. 1464286,9. 1464286,9. 1464286,9. 1464286,9. 1464286,9. 1464286,9. 1428566,9. 1428578,9. 1428574,9. 1428573,9. 1428572,9. 1428571,9. 1428571,9. 1428571,9. 1428571,9. 1428571,9. 1392786,9. 1392859,9. 1392862,9. 1392859,9. 1392858,9. 1392857,9. 1392857,9. 1392857,9. 1392857,9. 1392857,9. 1356968,9. 1357079,9. 959748 96474 969755 974781 979815 984854 989896 994944 94985 954432 959416 964451 969509 974579 979656 984736 98982 994907 949821 954227 95903 964106 969212 974333 979462 984592 989726 994863 949921 954266 135714,9.958808 1357144,9. 1357143,9. 1357143,9. 1357143,9. 1357143,9. 1357143,9. 1357143,9. 1321154,9. 1321291,9. 1321381,9. 1321424,9. 1321428,9. 1321428,9. 1321428,9. 1321429,9. 1321429,9. 1321429,9. 1285354,9. 1285506,9. l285624,9. 1285696,9. 1285711,9. 1285713,9. 1285714,9. 1285714,9. 1285714,9. 187 96377 968919 97409 979269 984451 989635 994818 950035 954357 958854 96358 968685 973894 979114 984338 989561 994783 95015 954483 958914 963531 968552 973781 979024 984272 989519 1140,0. 1141,0. 1142,0. 1143,0. 1144,0. 1145,0. 1146,0. 1147,0. 1148,0. 1149,0. 1150,0. 1151,0. 1152,0. 1153,0. 1154,0. 1155,0. 1156,0. 1157,0. 1158,0. 1159,0. 1160,0. 1161,0. 1162,0. 1163,0. 1164,0. 1165,0. 1166,0. 1167,0. 1168,0. 1169,0. 1170,0. 1171,0. 1172,0. 1173,0. 1174,0. 1175,0. 1176,0. 1177,0. 1178,0. 1179,0. 1180,0. 1181,0. 1182,0. 1183,0. 1184,0. 1185,0. 1186,0. 1187,0. 1188,0. 1189,0. 1190,0. 1191,0. 1192,0. 1193,0. 1194,0. 1195,0. 1196,0. 1285714,9. 1249564,9. 1249736,9. 994762 950305 954607 124987,9.959026 1249962,9. 1249993,9. 1249997,9. 1249999,9. 963605 968537 973766 979012 125,9.984262 125,9.989513 125,9.994761 1213804,9. 95039 121399,9.954689 1214131,9. 1214232,9. 1214274,9. 1214281,9. 1214284,9. 1214285,9. 1214285,9. 12l4286,9. 1178079,9. 1178272,9. 1178418,9. 1178515,9. 1178555,9. 1178564,9. 1178569,9. 959133 963729 968639 97385 979077 984312 989546 994776 950401 954721 959187 96386 968841 974016 979209 117857,9.984408 1178571,9. 1178571,9. 1142387,9. 1142585,9. 1142736,9. 1142813,9. 1142837,9. 1142848,9. 1142853,9. 1142856,9. 1142857,9. 1142857,9. 1106731,9. 1106919,9. 1107049,9. 1107097,9. 1107121,9. 1107133,9. 1107139,9. 110714l,9. 1107142,9. 1107143,9. 1071118,9. 1071263,9. 989609 994807 950388 954702 959165 964019 969112 974241 979386 98454 989695 994848 950401 954727 959373 96437 969418 974493 979585 984686 98979 994895 950452 954925 107135,9.959752 1071389,9. 96472 107l41,9.969725 107142,9.974739 1197,0. 1198,0. 1199,0. 1200,0. 1201,0. 1202,0. 1203,0. 1204,0. 1205,0. 1206,0. 1207,0. 1208,0. 1209,0. 1210,0. *Element, 1 I I I I ‘ ‘ OD\]O\U"I|I>UUN \D I 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 1, 293, 293, 294, 116, 305, 306, 115, 307, 308, 114, 309, 310, 113, 311, 312, 112, 313, 314, 111, 315, 316, 110, 317, 318, 109, 319, 320, 108, 321, 322, 107, 323, 324, 106, 325, 326, 105, 327, 328, 104, 329, 330, 1071425,9.979773 1071427,9.984821 1071428,9.989878 1071428,9.994938 103555,9.95049 1035628,9.955174 1035668,9.960047 1035691,9.964996 1035703,9.969962 1035709,9.974937 1035712,9.979918 1035713,9.984922 1035714,9.989943 1035714,9.994969 type=CPE4P 305, 116 294, 306, 305 2, 27, 306 305, 307, 115 306, 308, 307 27, 28, 308 307, 309, 114 308, 310, 309 28, 29, 310 309, 311, 113 310, 312, 311 29, 30, 312 311, 313, 112 312, 314, 313 30, 31, 314 313, 315, 111 314, 316, 315 31, 32, 316 315, 317, 110 316, 318, 317 32, 33, 318 317, 319, 109 318, 320, 319 33, 34, 320 319, 321, 108 320, 322, 321 34, 35, 322 321, 323, 107 322, 324, 323 35, 36, 324 323, 325, 106 324, 326, 325 36, 37, 326 325, 327, 105 326, 328, 327 37, 38, 328 327, 329, 104 328, 330, 329 38, 39, 330 329, 331, 103 330, 332, 331 39, 40, 332 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 103, 331, 333, 102 331, 332, 334, 333 332, 40, 41, 334 102, 333, 296, 4 333, 334, 295, 296 334, 41, 3, 295 71, 297, 335, 70 297, 298, 336, 335 298, 22, 23, 336 70, 335, 337, 69 335, 336, 338, 337 336, 23, 24, 338 69, 337, 339, 68 337, 338, 340, 339 338, 24, 25, 340 68, 339, 341, 67 339, 340, 342, 341 340, 25, 26, 342 67, 341, 293, 1 341, 342, 294, 293 342, 26, 2, 294 48, 299, 343, 47 299, 300, 344, 343 300, 95, 96, 344 47, 343, 345, 46 343, 344, 346, 345 344, 96, 97, 346 46, 345, 347, 45 345, 346, 348, 347 346, 97, 98, 348 45, 347, 349, 44 347, 348, 350, 349 348, 98, 99, 350 44, 349, 351, 43 349, 350, 352, 351 350, 99, 100, 352 43, 351, 353, 42 351, 352, 354, 353 352, 100, 101, 354 42, 353, 295, 3 353, 354, 296, 295 354, 101, 4, 296 22, 298, 355, 21 298, 297, 356, 355 297, 71, 72, 356 21, 355, 357, 20 355, 356, 358, 357 356, 72, 73, 358 20, 357, 359, 19 357, 358, 360, 359 358, 73, 74, 360 19, 359, 361, 18 359, 360, 362, 361 360, 74, 75, 362 18, 361, 363, 17 361, 362, 364, 363 362, 75, 76, 364 188 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 17, 363, 302, 6 363, 364, 301, 302 364, 76, 5, 301 95, 300, 365, 94 300, 299, 366, 365 299, 48, 49, 366 94, 365, 367, 93 365, 366, 368, 367 366, 49, 50, 368 93, 367, 369, 92 367, 368, 370, 369 368, 50, 51, 370 92, 369, 304, 8 369, 370, 303, 304 370, 51, 7, 303 8, 304, 371, 91 304, 303, 372, 371 303, 7, 52, 372 91, 371, 373, 90 371, 372, 374, 373 372, 52, 53, 374 90, 373, 375, 89 373, 374, 376, 375 374, 53, 54, 376 89, 375, 377, 88 375, 376, 378, 377 376, 54, 55, 378 88, 377, 379, 87 377, 378, 380, 379 378, 55, 56, 380 87, 379, 381, 86 379, 380, 382, 381 380, 56, 57, 382 86, 381, 383, 85 381, 382, 384, 383 382, 57, 58, 384 85, 383, 385, 84 383, 384, 386, 385 384, 58, 59, 386 84, 385, 387, 83 385, 386, 388, 387 386, 59, 60, 388 83, 387, 389, 82 387, 388, 390, 389 388, 60, 61, 390 82, 389, 391, 81 389, 390, 392, 391 390, 61, 62, 392 81, 391, 393, 80 391, 392, 394, 393 392, 62, 63, 394 80, 393, 395, 79 393, 394, 396, 395 394, 63, 64, 396 79, 395, 397, 78 395, 396, 398, 397 396, 64, 65, 398 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 78, 397, 399, 77 397, 398, 400, 399 398, 65, 66, 400 77, 399, 301, 5 399, 400, 302, 301 400, 66, 6, 302 9, 403, 417, 141 403, 404, 418, 417 404, 401, 419, 418 401, 405, 420, 419 405, 10, 152, 420 141, 417, 421, 140 417, 418, 422, 421 418, 419, 423, 422 419, 420, 424, 423 420, 152, 153, 424 140, 421, 425, 139 421, 422, 426, 425 422, 423, 427, 426 423, 424, 428, 427 424, 153, 154, 428 139, 425, 429, 138 425, 426, 430, 429 426, 427, 431, 430 427, 428, 432, 431 428, 154, 155, 432 138, 429, 433, 137 429, 430, 434, 433 430, 431, 435, 434 431, 432, 436, 435 432, 155, 156, 436 137, 433, 437, 136 433, 434, 438, 437 434, 435, 439, 438 435, 436, 440, 439 436, 156, 157, 440 136, 437, 441, 135 437, 438, 442, 441 438, 439, 443, 442 439, 440, 444, 443 440, 157, 158, 444 135, 441, 445, 134 441, 442, 446, 445 442, 443, 447, 446 443, 444, 448, 447 444, 158, 159, 448 134, 445, 449, 133 445, 446, 450, 449 446, 447, 451, 450 447, 448, 452, 451 448, 159, 160, 452 133, 449, 453, 132 449, 450, 454, 453 450, 451, 455, 454 451, 452, 456, 455 452, 160, 161, 456 132, 453, 457, 131 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 453, 454, 455, 456, 131, 457, 458, 459, 460, 130, 461, 462, 463, 464, 129, 465, 466, 467, 468, 128, 469, 470, 471, 472, 127, 473, 474, 475, 476, 145, 146, 409, 410, 411, 412, 478, 482, 483, 484, 403, 479, 480, 481, 405, 488, 487, 486, 409, 10, 151, 150, 149, 120, 413, 414, 415, 416, 454, 458, 457 455, 459, 458 456, 460, 459 161, 162, 460 457, 461, 130 458, 462, 461 459, 463, 462 460, 464, 463 162, 163, 464 461, 465, 129 462, 466, 465 463, 467, 466 464, 468, 467 163, 164, 468 465, 469, 128 466, 470, 469 467, 471, 470 468, 472, 471 164, 165, 472 469, 473, 127 470, 474, 473 471, 475, 474 472, 476, 475 165, 166, 476 473, 407, 12 474, 406, 407 475, 402, 406 476, 408, 402 166, 11, 408 146, 478, 477 147, 409, 478 410, 482, 478 411, 483, 482 412, 484, 483 401, 404, 484 482, 481, 477 483, 480, 481 484, 479, 480 404, 403, 479 9, 142, 479 142, 143, 480 143, 144, 481 144, 145, 477 401, 412, 488 412, 411, 487 411, 410, 486 410, 409, 485 147, 148, 485 405, 488, 151 488, 487, 150 487, 486, 149 486, 485, 148 121, 413, 489 414, 492, 489 415, 491, 492 416, 490, 491 402, 408, 490 189 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 327, 408, 11, 117, 490 490, 117, 118, 491 491, 118, 119, 492 492, 119, 120, 489 407, 406, 498, 497 406, 402, 416, 498 497, 498, 499, 496 498, 416, 415, 499 496, 499, 500, 495 499, 415, 414, 500 495, 500, 494, 493 500, 414, 413, 494 413, 121, 122, 494 494, 122, 123, 493 123, 124, 495, 493 124, 125, 496, 495 125, 126, 497, 496 126, 12, 407, 497 22, 121, 120, 23 23, 120, 119, 24 24, 119, 118, 25 25, 118, 117, 26 26, 117, 11, 2 121, 22, 21, 122 122, 21, 20, 123 123, 20, 19, 124 124, 19, 18, 125 125, 18, 17, 126 126, 17, 6, 12 2, 11, 166, 27 27, 166, 165, 28 28, 165, 164, 29 29, 164, 163, 30 30, 163, 162, 31 31, 162, 161, 32 32, 161, 160, 33 33, 160, 159, 34 34, 159, 158, 35 35, 158, 157, 36 36, 157, 156, 37 37, 156, 155, 38 38, 155, 154, 39 39, 154, 153, 40 40, 153, 152, 41 41, 152, 10, 3 12, 6, 66, 127 127, 66, 65, 128 128, 65, 64, 129 129, 64, 63, 130 130, 63, 62, 131 131, 62, 61, 132 132, 61, 60, 133 133, 60, 59, 134 134, 59, 58, 135 135, 58, 57, 136 136, 57, 56, 137 137, 56, 55, 138 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 340, 341, 342, 343, 344, 345, 346, 347, 348, 349, 350, 351, 352, 353, 354, 355, 356, 357, 358, 359, 360, 361, 362, 363, 364, 365, 366, 367, 368, 369, 370, 371, 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384, 138, 139, 140, 141, 149, 150, 151, 49, 50, 51, 144, 145, 146, 147, 148, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 506, 571, 572, 573, 574, 575, 576, 577, 578, 579, 580, 505, 581, 582, 583, 584, 585, 586, 587, 588, 589, 590, 504, 591, 592, 593, 594, 595, 596, 597, 598, 55, 54, 53, 52, 44, 43, 42, 144, 143, 142, 49, 48, 47, 46, 45, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 13, 571, 572, 573, 574, 575, 576, 577, 578, 579, 580, 167, 581, 582, 583, 584, 585, 586, 587, 588, 589, 590, 168, 591, 592, 593, 594, 595, 596, 597, 598, 599, 54, 139 53, 140 52, 141 7, 9 43, 150 42, 151 3, 10 143, 50 142, 51 9, 7 48, 145 47, 146 46, 147 45, 148 44, 149 571, 506 572, 571 573, 572 574, 573 575, 574 576, 575 577, 576 578, 577 579, 578 580, 579 167, 580 581, 505 582, 581 583, 582 584, 583 585, 584 586, 585 587, 586 588, 587 589, 588 590, 589 168, 590 591, 504 592, 591 593, 592 594, 593 595, 594 596, 595 597, 596 598, 597 599, 598 600, 599 169, 600 601, 503 602, 601 603, 602 604, 603 605, 604 606, 605 607, 606 608, 607 609, 608 385, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396, 397, 398, 399, 400, 401, 402, 403, 404, 405, 406, 407, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, 431, 432, 433, 434, 435, 436, 437, 438, 439, 440, 441, 599, 600, 503, 601, 602, 603, 604, 605, 606, 607, 608, 609, 610, 501, 611, 612, 613, 614, 615, 616, 617, 618, 619, 620, 510, 621, 622, 623, 624, 625, 626, 627, 628, 629, 630, 509, 631, 632, 633, 634, 635, 636, 637, 638, 639, 640, 508, 641, 642, 643, 644, 645, 646, 647, 648, 649, 650, 600, 169, 601, 602, 603, 604, 605, 606, 607, 608, 609, 610, 170, 611, 612, 613, 614, 615, 616, 617, 618, 619, 620, 171, 621, 622, 623, 624, 625, 626, 627, 628, 629, 630, 172, 631, 632, 633, 634, 635, 636, 637, 638, 639, 640, 173, 641, 642, 643, 644, 645, 646, 647, 648, 649, 650, 174, 190 610, 170, 611, 612, 613, 614, 615, 616, 617, 618, 619, 620, 171, 621, 622, 623, 624, 625, 626, 627, 628, 629, 630, 172, 631, 632, 633, 634, 635, 636, 637, 638, 639, 640, 173, 641, 642, 643, 644, 645, 646, 647, 648, 649, 650, 174, 651, 652, 653, 654, 655, 656, 657, 658, 659, 660, 175, 609 610 501 611 612 613 614 615 616 617 618 619 620 510 621 622 623 624 625 626 627 628 629 630 509 631 632 633 634 635 636 637 638 639 640 508 641 642 643 644 645 646 647 648 649 650 507 651 652 653 654 655 656 657 658 659 660 442, 443, 444, 445, 446, 447, 448, 449, 450, 451, 452, 453, 454, 455, 456, 457, 458, 459, 460, 461, 462, 463, 464, 465, 466, 467, 468, 469, 470, 471, 472, 473, 474, 475, 476, 477, 478, 479, 480, 481, 482, 483, 484, 485, 486, 487, 488, 489, 490, 491, 492, 493, 494, 495, 496, 497, 498, 507, 651, 652, 653, 654, 655, 656, 657, 658, 659, 660, 279, 280, 281, 664, 674, 675, 505, 504, 503, 662, 676, 677, 663, 678, 679, 513, 514, 515, 516, 517, 518, 665, 680, 681, 682, 683, 684, 666, 685, 686, 687, 688, 689, 69, 68, 67, 667, 690, 691, 668, 692, 693, 669, 694, 695, 670, 651, 652, 653, 654, 655, 656, 657, 658, 659, 660, 175, 280, 281, 282, 674, 675, 506, 504, 503, 501, 676, 677, 511, 678, 679, 512, 514, 515, 516, 517, 518, 72, 680, 681, 682, 683, 684, 71, 685, 686, 687, 688, 689, 70, 68, 67, 11 690, 691, 519, 692, 693, 520, 694, 695, 521, 696, 690, 691, 519, 185, 184, 183, 182, 181, 180, 179, 178, 177, 176, 14, 674, 675, 506, 663, 662, 505, 676, 677, 511, 678, 679, 512, 666, 665, 513, 680, 681, 682, 683, 684, 71, 685, 686, 687, 688, 689, 70, 671, 670, 669, 668, 667, 69, 692, 693, 520, 694, 695, 521, 696, 697, 522, 698, 186 185 184 183 182 181 180 179 178 177 176 664 674 675 661 663 662 662 676 677 663 678 679 661 666 665 665 680 681 682 683 684 666 685 686 687 688 689 661 671 670 669 668 667 667 690 691 668 692 693 669 694 695 670 696 697 671 499, 500, 501, 502, 503, 504, 505, 506, 507, 508, 509, 510, 511, 512, 513, 514, 515, 516, 517, 518, 519, 520, 521, 522, 523, 524, 525, 526, 527, 528, 529, 530, 531, 532, 533, 534, 535, 536, 537, 538, 539, 540, 541, 542, 543, 544, 545, 546, 547, 548, 549, 550, 551, 552, 553, 554, 555, 696, 697, 671, 698, 699, 524, 525, 672, 700, 673, 701, 514, 513, 512, 511, 706, 715, 716, 717, 509, 508, 507, 703, 718, 719, 704, 720, 721, 705, 722, 723, 190, 191, 192, 707, 724, 725, 708, 726, 727, 528, 529, 530, 531, 532, 709, 728, 729, 730, 731, 710, 732, 733, 734, 735, 74, 73, 697, 699, 698 522, 523, 699 698, 673, 661 699, 672, 673 523, 524, 672 525, 700, 672 276, 277, 700 700, 701, 673 277, 278, 701 701, 664, 661 278, 279, 664 513, 715, 706 512, 716, 715 511, 717, 716 501, 510, 717 715, 705, 702 716, 704, 705 717, 703, 704 510, 509, 703 508, 718, 703 507, 719, 718 186, 187, 719 718, 720, 704 719, 721, 720 187, 188, 721 720, 722, 705 721, 723, 722 188, 189, 723 722, 708, 702 723, 707, 708 189, 190, 707 191, 724, 707 192, 725, 724 193, 526, 725 724, 726, 708 725, 727, 726 526, 527, 727 726, 710, 702 727, 709, 710 527, 528, 709 529, 728, 709 530, 729, 728 531, 730, 729 532, 731, 730 5, 76, 731 728, 732, 710 729, 733, 732 730, 734, 733 731, 735, 734 76, 75, 735 732, 714, 702 733, 713, 714 734, 712, 713 735, 711, 712 75, 74, 711 73, 736, 711 72, 518, 736 556, 557, 558, 559, 560, 561, 562, 563, 564, 565, 566, 567, 568, 569, 570, 571, 572, 573, 574, 575, 576, 577, 578, 579, 580, 581, 582, 583, 584, 585, 586, 587, 588, 589, 590, 591, 592, 593, 594, 595, 596, 597, 598, 599, 600, 601, 602, 603, 604, 605, 606, 607, 608, 609, 610, 611, 612, 711, 736, 712, 737, 713, 738, 714, 739, 276, 525, 524, 523, 522, 521, 520, 519, 275, 740, 741, 742, 743, 744, 745, 746, 274, 747, 748, 749, 750, 751, 752, 753, 273, 754, 755, 756, 757, 758, 759, 760, 272, 761, 762, 763, 764, 765, 766, 767, 271, 768, 769, 770, 771, 772, 773, 774, 270, 736, 737, 712 518, 517, 737 737, 738, 713 517, 516, 738 738, 739, 714 516, 515, 739 739, 706, 702 515, 514, 706 525, 740, 275 524, 741, 740 523, 742, 741 522, 743, 742 521, 744, 743 520, 745, 744 519, 746, 745 1, 116, 746 740, 747, 274 741, 748, 747 742, 749, 748 743, 750, 749 744, 751, 750 745, 752, 751 746, 753, 752 116, 115, 753 747, 754, 273 748, 755, 754 749, 756, 755 750, 757, 756 751, 758, 757 752, 759, 758 753, 760, 759 115, 114, 760 754, 761, 272 755, 762, 761 756, 763, 762 757, 764, 763 758, 765, 764 759, 766, 765 760, 767, 766 114, 113, 767 761, 768, 271 762, 769, 768 763, 770, 769 764, 771, 770 765, 772, 771 766, 773, 772 767, 774, 773 113, 112, 774 768, 775, 270 769, 776, 775 770, 777, 776 771, 778, 777 772, 779, 778 773, 780, 779 774, 781, 780 112, 111, 781 775, 782, 269 191 613, 614, 615, 616, 617, 618, 619, 620, 621, 622, 623, 624, 625, 626, 627, 628, 629, 630, 631, 632, 633, 634, 635, 636, 637, 638, 639, 640, 641, 642, 643, 644, 645, 646, 647, 648, 649, 650, 651, 652, 653, 654, 655, 656, 657, 658, 659, 660, 661, 662, 663, 664, 665, 666, 667, 668, 669, 775, 776, 777, 778, 779, 780, 781, 269, 782, 783, 784, 785, 786, 787, 788, 268, 789, 790, 791, 792, 793, 794, 795, 267, 796, 797, 798, 799, 800, 801, 802, 266, 803, 804, 805, 806, 807, 808, 809, 265, 810, 811, 812, 813, 814, 815, 816, 264, 817, 818, 819, 820, 821, 822, 823, 263, 824, 776, 777, 778, 779, 780, 781, 111, 782, 783, 784, 785, 786, 787, 788, 110, 789, 790, 791, 792, 793, 794, 795, 109, 796, 797, 798, 799, 800, 801, 802, 108, 803, 804, 805, 806, 807, 808, 809, 107, 810, 811, 812, 813, 814, 815, 816, 106, 817, 818, 819, 820, 821, 822, 823, 105, 824, 825, 783, 784, 785, 786, 787, 788, 110, 789, 790, 791, 792, 793, 794, 795, 109, 796, 797, 798, 799, 800, 801, 802, 108, 803, 804, 805, 806, 807, 808, 809, 107, 810, 811, 812, 813, 814, 815, 816, 106, 817, 818, 819, 820, 821, 822, 823, 105, 824, 825, 826, 827, 828, 829, 830, 104, 831, 832, 782 783 784 785 786 787 788 268 789 790 791 792 793 794 795 267 796 797 798 799 800 801 802 266 803 804 805 806 807 808 809 265 810 811 812 813 814 815 816 264 817 818 819 820 821 822 823 263 824 825 826 827 828 829 830 262 831 670, 671, 672, 673, 674, 675, 676, 677, 678, 679, 680, 681, 682, 683, 684, 685, 686, 687, 688, 689, 690, 691, 692, 693, 694, 695, 696, 697, 698, 699, 700, 701, 702, 703, 704, 705, 706, 707, 708, 709, 710, 711, 712, 713, 714, 715, 716, 717, 718, 719, 720, 721, 722, 723, 724, 725, 726, 825, 826, 827, 828, 829, 830, 262, 831, 832, 833, 834, 835, 836, 837, 261, 838, 839, 840, 841, 842, 843, 844, 209, 540, 541, 542, 543, 544, 545, 546, 208, 845, 846, 847, 848, 849, 850, 851, 207, 852, 853, 854, 855, 856, 857, 858, 206, 859, 860, 861, 862, 863, 864, 865, 205, 866, 867, 826, 833, 832 827, 834, 833 828, 835, 834 829, 836, 835 830, 837, 836 104, 103, 837 831, 838, 261 832, 839, 838 833, 840, 839 834, 841, 840 835, 842, 841 836, 843, 842 837, 844, 843 103, 102, 844 838, 539, 260 839, 538, 539 840, 537, 538 841, 536, 537 842, 535, 536 843, 534, 535 844, 533, 534 102, 4, 533 540, 845, 208 541, 846, 845 542, 847, 846 543, 848, 847 544, 849, 848 545, 850, 849 546, 851, 850 8, 91, 851 845, 852, 207 846, 853, 852 847, 854, 853 848, 855, 854 849, 856, 855 850, 857, 856 851, 858, 857 91, 90, 858 852, 859, 206 853, 860, 859 854, 861, 860 855, 862, 861 856, 863, 862 857, 864, 863 858, 865, 864 90, 89, 865 859, 866, 205 860, 867, 866 861, 868, 867 862, 869, 868 863, 870, 869 864, 871, 870 865, 872, 871 89, 88, 872 866, 873, 204 867, 874, 873 868, 875, 874 727, 728, 729, 730, 731, 732, 733, 734, 735, 736, 737, 738, 739, 740, 741, 742, 743, 744, 745, 746, 747, 748, 749, 750, 751, 752, 753, 754, 755, 756, 757, 758, 759, 760, 761, 762, 763, 764, 765, 766, 767, 768, 769, 770, 771, 772, 773, 774, 775, 776, 777, 778, 779, 780, 781, 782, 783, 868, 869, 870, 871, 872, 204, 873, 874, 875, 876, 877, 878, 879, 203, 880, 881, 882, 883, 884, 885, 886, 202, 887, 888, 889, 890, 891, 892, 893, 201, 894, 895, 896, 897, 898, 899, 900, 200, 901, 902, 903, 904, 905, 906, 907, 199, 908, 909, 910, 911, 912, 913, 914, 198, 915, 916, 917, 869, 870, 871, 872, 88, 873, 874, 875, 876, 877, 878, 879, 87, 880, 881, 882, 883, 884, 885, 886, 86, 887, 888, 889, 890, 891, 892, 893, 85, 894, 895, 896, 897, 898, 899, 900, 84, 901, 902, 903, 904, 905, 906, 907, 83, 908, 909, 910, 911, 912, 913, 914, 82, 915, 916, 917, 918, 192 87, 86, 85, 84, 83, 82, 81, 876, 877, 878, 879, 880, 881, 882, 883, 884, 885, 886, 887, 888, 889, 890, 891, 892, 893, 894, 895, 896, 897, 898, 899, 900, 901, 902, 903, 904, 905, 906, 907, 908, 909, 910, 911, 912, 913, 914, 915, 916, 917, 918, 919, 920, 921, 922, 923, 924, 925, 875 876 877 878 879 203 880 881 882 883 884 885 886 202 887 888 889 890 891 892 893 201 894 895 896 897 898 899 900 200 901 902 903 904 905 906 907 199 908 909 910 911 912 913 914 198 915 916 917 918 919 920 921 197 922 923 924 784, 785, 786, 787, 788, 789, 790, 791, 792, 793, 794, 795, 796, 797, 798, 799, 800, 801, 802, 803, 804, 805, 806, 807, 808, 809, 810, 811, 812, 813, 814, 815, 816, 817, 818, 819, 820, 821, 822, 823, 824, 825, 826, 827, 828, 829, 830, 831, 832, 833, 834, 835, 836, 837, 838, 839, 840, 918, 919, 920, 921, 197, 922, 923, 924, 925, 926, 927, 928, 196, 929, 930, 931, 932, 933, 934, 935, 195, 936, 937, 938, 939, 940, 941, 942, 194, 943, 944, 945, 946, 947, 948, 949, 551, 552, 553, 554, 555, 556, 557, 558, 960, 967, 968, 969, 970, 971, 972, 973, 959, 974, 975, 976, 977, 919, 920, 921, 81, 922, 923, 924, 925, 926, 927, 928, 80, 929, 930, 931, 932, 933, 934, 935, 79, 936, 937, 938, 939, 940, 941, 942, 78, 943, 944, 945, 946, 947, 948, 949, 77, 552, 553, 554, 555, 556, 557, 558, 502, 967, 968, 969, 970, 971, 972, 973, 559, 974, 975, 976, 977, 978, 926, 927, 928, 80, 929, 930, 931, 932, 933, 934, 935, 79, 936, 937, 938, 939, 940, 941, 942, 78, 943, 944, 945, 946, 947, 948, 949, 77, 526, 527, 528, 529, 530, 531, 532, 5, 967, 968, 969, 970, 971, 972, 973, 559, 974, 975, 976, 977, 978, 979, 980, 560, 981, 982, 983, 984, 985, 532 925 926 927 928 196 929 930 931 932 933 934 935 195 936 937 938 939 940 941 942 194 943 944 945 946 947 948 949 193 526 527 528 529 530 531 960 967 968 969 970 971 972 973 959 974 975 976 977 978 979 980 958 981 982 983 984 841, 978, 979, 986, 985 842, 979, 980, 987, 986 843, 980, 560, 561, 987 844, 958, 981, 957, 950 845, 981, 982, 956, 957 846, 982, 983, 955, 956 847, 983, 984, 954, 955 848, 984, 985, 953, 954 849, 985, 986, 952, 953 850, 986, 987, 951, 952 851, 987, 561, 562, 951 852, 562, 563, 988, 951 853, 563, 564, 989, 988 854, 564, 251, 252, 989 855, 951, 988, 990, 952 856, 988, 989, 991, 990 857, 989, 252, 253, 991 858, 952, 990, 992, 953 859, 990, 991, 993, 992 860, 991, 253, 254, 993 861, 953, 992, 994, 954 862, 992, 993, 995, 994 863, 993, 254, 255, 995 864, 954, 994, 996, 955 865, 994, 995, 997, 996 866, 995, 255, 256, 997 867, 955, 996, 998, 956 868, 996, 997, 999, 998 869, 997, 256, 257, 999 870,956,998,1000,957 871,998,999,1001,1000 872,999,257,258,1001 873,957,1000,962,950 874,1000,1001,961,962 875,1001,258,259,961 876,259,260,539,961 877,961,539,538,962 878,962,538,537,950 879,537,536,966,950 880,536,535,965,966 881,535,534,964,965 882,534,533,963,964 883,533,4,101,963 884,101,100,1002,963 885,100,99,1003,1002 886,99,98,1004,1003 887,98,97,547,1004 888,963,1002,1005,964 889,1002,1003,1006,1005 890,1003,1004,1007,1006 891,1004,547,548,1007 892,964,1005,1008,965 893,1005,1006,1009,1008 894,1006,1007,1010,1009 895,1007,548,549,1010 896,965,1008,1011,966 897,1008,1009,1012,1011 898,1009,1010,1013,1012 899,1010,549,550,1013 900,966,101l,958,950 901,1011,1012,959,958 902,1012,1013,960,959 903,1013,550,551,960 904,210,211,1031,1021 905,211,212,1032,1031 906,212,213,1033,1032 907,213,214,1034,1033 908,214,215,1035,1034 909,215,216,1036,1035 910,216,217,565,1036 911,1021,1031,1020,1014 912,1031,1032,1019,1020 913,1032,1033,1018,1019 914,1033,1034,1017,1018 915,1034,1035,1016,1017 916,1035,1036,1015,1016 917,1036,565,566,1015 918,566,567,1037,1015 919,567,568,1038,1037 920,568,569,1039,1038 921,569,570,1040,1039 922,570,502,558,1040 923,1015,1037,1041,1016 924,1037,1038,1042,1041 925,1038,1039,1043,1042 926,1039,1040,1044,1043 927,1040,558,557,1044 928,1016,1041,1045,1017 929,1041,1042,1046,1045 930,1042,1043,1047,1046 931,1043,1044,1048,1047 932,1044,557,556,1048 933,1017,1045,1049,1018 934,1045,1046,1050,1049 935,1046,1047,1051,1050 936,1047,1048,1052,1051 937,1048,556,555,1052 938,1018,1049,1053,1019 939,1049,1050,1054,1053 940,1050,1051,1055,1054 941,1051,1052,1056,1055 942,1052,555,554,1056 943,1019,1053,1057,1020 944,1053,1054,1058,1057 945,1054,1055,1059,1058 946,1055,1056,1060,1059 947,1056,554,553,1060 948,1020,1057,1025,1014 949,1057,1058,1024,1025 950,1058,1059,1023,1024 951,1059,1060,1022,1023 952,1060,553,552,1022 953,552,551,1061,1022 954,551,550,1062,1061 193 955,550,549,1063,1062 956,549,548,1064,1063 957,548,547,1065,1064 958,547,97,96,1065 959,1022,1061,1066,1023 960,1061,1062,1067,1066 961,1062,1063,1068,1067 962,1063,1064,1069,1068 963,1064,1065,1070,1069 964,1065,96,95,1070 965,1023,1066,1071,1024 966,1066,1067,1072,1071 967,1067,1068,1073,1072 968,1068,1069,1074,1073 969,1069,1070,1075,1074 970,1070,95,94,1075 971,1024,1071,1076,1025 972,1071,1072,1077,1076 973,1072,1073,1078,1077 974,1073,1074,1079,1078 975,1074,1075,1080,1079 976,1075,94,93,1080 977,1025,1076,1030,1014 978,1076,1077,1029,1030 979,1077,1078,1028,1029 980,1078,1079,1027,1028 981,1079,1080,1026,1027 982,1080,93,92,1026 983,92,8,546,1026 984,1026,546,545,1027 985,1027,545,544,1028 986,1028,544,543,1029 987,1029,543,542,1030 988,1030,542,541,1014 989,541,540,1021,1014 990,540,209,210,1021 99l,217,218,1081,565 992,218,219,1082,1081 993,219,220,1083,1082 994,220,221,1084,1083 995,221,222,1085,1084 996,222,223,1086,1085 997,223,224,1087,1086 998,224,225,1088,1087 999,225,226,1089,1088 1000,226,227,1090,1089 1001,227,15,228,1090 1002,565,1081,1091,566 1003,1081,1082,1092,1091 1004,1082,1083,1093,1092 1005,1083,1084,1094,1093 1006,1084,1085,1095,1094 1007,1085,1086,1096,1095 1008,1086,1087,1097,1096 1009,1087,1088,1098,1097 1010,1088,1089,1099,1098 1011,1089,1090,1100,1099 1012,1090,228,229,1100 1013,566,109l,1101,567 1014,1091. 1015,1092. 1016,1093. 1017,1094. 1018,1095. 1019,1096. 1020,1097. 1021,1098. 1022,1099. 1092. 1093. 1094. 1095. 1096. 1097 1098. 1099. 1100. 1102,1101 1103,1102 1104,1103 1105,1104 1106,1105 .1107,1106 1108,1107 1109,1108 1110,1109 1023,1100,229,230,1110 1024,567,1101,1111,568 1025,1101. 1026,1102. 1027,1103. 1028,1104. 1029,1105, 1030,1106. 1031,1107. 1032,1108. 1033,1109, 1102. 1103. 1104 1105. 1106. 1107. 1108 1109. 1110. .1114. .1118. 1112. 1113. 1111 1112 1113 1114 1115 1116 1117 1118 1119 1115. 1116. 1117. 1119. 1120. 1034,1110,230,231,1120 1035,568,1111,1121,569 1036,1111. 1037,1112, 1038,1113. 1039,1114. 1040,1115. 1041,1116. 1042,1117. 1043,1118. 1044,1119. 1045,1120. 1047,1121. 1048,1122. 1049,1123. 1050,1124. 1051,1125. 1052,1126. 1053,1127. 1054,1128. 1055,1129. 1056,1130. 1058,1131. 1059,1132. 1060,1133. 1061,1134. 1062,1135. 1063,1136. 1064,1137. 1065,1138. 1066,1139. 1067,1140. 1112. 1113. 1114 1115. 1116. 1117 1118. 1119. 1120. .1124. .1127. 1122. 1123. 1121 1122 1123 1124 1125 1126 1127 1128 1129 1125. 1126. 1128. 1129. 1130. 231,232,1130 1046,569,1121,1131,570 1122. 1123. 1124 1125. 1126. 1127 1128. 1129. 1130. 1132,1131 1133,1132 .1134,1133 1135,1134 1136,1135 .1137.1136 1138,1137 1139,1138 1140,1139 232.233.1140 1057,570,1131,1141,502 1132. 1133. 1134. 1135. 1136. 1137. 1138. 1139. 1140. 1142,1141 1143,1142 1144,1143 1145,1144 1146,1145 1147,1146 1148,1147 1149,1148 1150,1149 233,234,1150 1068,502,1141,1151,559 1069. 1070. 1071. 1072. 1073. 1074. 1075. 1076. 1077, 1078. 1079. 1080. 1081. 1082. 1083. 1084. 1085. 1086. 1087. 1088. 1089. 1090. 1091. 1092. 1093. 1094. 1095. 1096. 1097. 1098. 1099. 1100. 1101. 1102. 1103. 1104. 1105. 1106. 1107. 1108. 1109. 1110. 1111. 1112. 1113. 1114. 1115. 1116. 1117. 1118. 1119. 1120. 1121. 1122. 1123. 1124. 1125. 1141. 1142. 1143. 1144. 1145. 1146. 1147. 1148. 1149. 1150. 1142. 1143. 1144 1145. 1146. 1147. 1148. 1149. 1150. .1154. 1152. 1153. 1151 1152 1153 1154 1155 1156 1157 1158 1159 1155. 1156. 1157. 1158. 1159. 1160. 234,235,1160 559,1151,1161,560 1151. 1152. 1153. 1154. 1155. 1156. 1157,1158. 1158,1159. 1159,1160. 1152. 1153. 1154. 1156. 1155. 1157. 1162,1161 1163,1162 1164,1163 1165,1164 1166,1165 1167,1166 1168,1167 1169,1168 1170,1169 1160,235,236,1170 560,1161,1171.561 1161. 1162. 1163. 1164. 1165. 1166. 1167. 1168. 1169. 1170. 1162. 1163. 1164. 1165. 1166. 1167, 1168. 1169. 1170. 1172,1171 1173,1172 1174,1173 1175,1174 1176,1175 1177,1176 1178,1177 1179,1178 1180,1179 236,237,1180 561,1171,1181,562 1171. 1172. 1173. 1174. 1175,1176 1176,1177. 1177,1178. 1178,1179. 1179,1180. 1172. 1173. 1174. 1175. 1182,1181 1183,1182 1184,1183 1185,1184 .1186,1185 1187,1186 1188,1187 1189,1188 1190,1189 1180,237,238,1190 562,1181,1191,563 1181. 1182. 1183. 1184. 1185. 1186. 1187. 1188. 1189. 1190. 1182. 1183. 1184. 1185. 1186. 1187. 1188. 1189. 1190. 1192,1191 1193,1192 1194,1193 1195,1194 1196,1195 1197,1196 1198,1197 1199,1198 1200,1199 238,239,1200 563,1191,1201,564 1191,1192. 1192,1193, 194 1202,1201 1203,1202 1126,1193,1194,1204,1203 1127,1194,1195,1205,1204 1128,1195,1196,1206,1205 1129,1196,1197,1207,1206 1130,1197,1198,1208,1207 1131,1198,1199,1209,1208 1132,1199,1200,1210,1209 1133,1200,239,240,1210 1134,564,1201,250,251 1135,1201,1202,249,250 1136,1202,1203,248,249 1137,1203,1204,247,248 1138,1204,1205,246,247 1139,1205,1206,245,246 ll40,1206,1207,244,245 1141,1207,1208,243,244 1142,1208,1209,242,243 1143,1209,1210,241,242 1144,1210,240,16,241 *Element, type=SPRINGA, Elset=MedLow 1145, 13, 16 *Element, type=SPRINGA, Elset=Low 1146, 14, *Spring, nonlinear 15 Elset=MedLow, O, O 0, 0.0032 400, 20 *Spring, nonlinear Elset=Low, 0,0 0, 0.0006 400, 20 *Orientation,name=0ri-3 1., 0., 0., 0., 1., 0. 2, 0. ** Region: (PCM- Section:Picked), (Material OrientationzPicked) *Elset, elset=_I1, generate 1, 162, 1 ** Section: PCM-Section *Solid Section, elset=_I1, orientation=Ori-3, material=YINPCM 1., ** Region: (Cell— Section:Picked), (Material Orientation:Picked) *Elset, elset=_12, generate 163, 288, 1 ** Section: Cell— Section *Solid Section, elset=_I2, orientation=Ori—3, materia1=YINCELL 1., ** Region: (membrane- sectionzPicked), (Material OrientationzPicked) *Elset, elset=_I3, generate 289, 342, 1 ** Section: membrane— section *Solid Section, elset=_I3, orientation=Ori-3, material=YINMEM 1., ** Region: (ECM— Section:Picked), (Material OrientationzPicked) *Elset, elset=_I4, generate 343, 1144, 1 ** Section: ECM-Section *Solid Section, elset=_I4, orientation=Ori—3, material=YINMatrix elset=shearstress 1., *Elset, 289, 290, 293, 294, 297, 298, 301, 302, 305, 306, 309, 310, 313, 314, 317, 318, 321, 322, 324, 325, 328, 329, 332, 333, 336, 337, 340, 341, 6, 9, 21, 24, 291, 295, 299, 303, 307, 311, 315, 319, 323, 326, 330, 334, 338, 342, 12, 27, 292, 296, 300, 304 308, 312, 316, 320 323, 327, 331, 335 339, 3. 15, 30, 18, 33, 36, 39, 42, 45, 48, 51, 54, 57, 60, 63, 64, 67, 70, 73, 76, 79, 82, 85, 88, 91, 94, 97, 100, 105, 108, 111, 114, 117, 120, 123, 126, 129, 132, 135, 138, 141, 144, 147, 150, 153, 156, 159, 162 *Nset, nset=NSUB, generate 1, 1210, 1 *Elset, elset=NSUB, generate 1, 1144, 1 *Nset, nset=_bottom, generate 13, 14, 1 *Nset, nset=_PickedSet76 13, 14, 15, 16, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292 *Elset, elset=_PickedSet76 195 343, 344, 347, 348, 351, 352, 375, 386, 419, 430, 443, 444, 447, 448, 451, 452, 455, 505, 520, 523, 530, 531, 572, 580, 604, 612, 636, 644, 668, 676, 700, 708, 732, 740, 764, 772, 796, 804, 857, 860, 869, 872, 904, 905, 908, 909, 991, 992, 995, 996, 999, 1000, 1023, 1034, 1067, 1078, 1111, 1122, 1135, 1136, 1139, 1140, 1143, 1144 *Submodel, 345, 349, 353, 397, 441, 445, 449, 453, 507, 526, 532, 588, 620, 652, 684, 716, 748, 780, 812, 863, 875, 906, 910, 993, 997, 1001, 1045, 1089, 1133, 1137, 1141, 346, 350, 364, 408, 442, 446, 450, 454, 509, 529, 564, 596, 628, 660, 692, 724, 756, 788, 854, 866, 876, 907, 990, 994, 998, 1012, 1056, 1100, 1134, 1138, 1142, exteriorTolerance=0.01 ** _PickedSet76, ** MATERIALS *Material, name=YINCELL *Elastic 0.5, 0.069 *Permeability, specific=9.81e~06 4.5e-09,2. *Material, *Elastic 1., 0.49 *Permeability, Specific=9.81e—O6 5e-14,2. *Material, name=YINMatrix *Elastic, type=ENGINEERING CONSTANTS 0.0457, 1., 0.0457, 0.07769, 0.7, 1.7, 0.5, 0.013441, 0.5, name=YINMEM *Permeability, specific=9.81e-O6 3.1392e-09,2. *Material, name=YINPCM *Elastic 1., 0.49 *Permeability, specific=9.81e—06 4e-09,2. *Material, name=membrane *Elastic, type=ENGINEERING CONSTANTS 1., 70., 1., 0.01, 0.3, 0.7, 5., 0.385, 5., *Permeability, specific=9.81e-06 9.81e—08,2. *INITIAL CONDITIONS, TYPE=RATIO NSUB, 2. *Boundary _bottom, 3, 3 ** ** 1% strain at 2% strain/minute ** STEP: Step—1 ** *Step, nlgeom, inc=1000 *Soils, consolidation, end=PERIOD 5., 30., , , *El Print, elset=shearstress, FREQUENCY=30 COORD, FLVEL *CONTROLS, PARAMETERS=FIELD, FIELD=PORE FLUID PRESSURE ,,0.001 ** name=Step-l, amplitude=RAMP, ** _____________________ ** ** BOUNDARY CONDITIONS ** ** Name: Submodel *Boundary, submodel, step=1, timescale _PickedSet76, 1, 1 _PickedSet76, 2, 2 Sub Type: _PickedSet76, 8, 8 ** ** OUTPUT REQUESTS ** *Restart, write, frequency=1 ** ** FIELD OUTPUT: F— Output-1 ** *Output, field *Node Output U, RF, POR *Element Output S, LE, VOIDR, SAT, FLVEL, COORD ** ** HISTORY OUTPUT: H- Output-1 ** *Output, history, variab1e=PRESELECT *El Print, freq=999999 *Node Print, freq=999999 *FILE FORMAT, INCREMENT *End Step ZERO B.6 Submodel for 1% strain at 20% strain/min ** ** .................... ** ** STEP: Step—l ** *Step, name=Step-1, nlgeom, amplitude=RAMP, inc=1000 *Soils, consolidation, end=PERIOD 0.1, 3., , , *El Print, elset=shearstress, FREQUENCY;3O COORD, FLVEL *CONTROLS, PARAMETERS=FIELD, FIELD=PORE FLUID PRESSURE 196 B.7 Submodel for 3% strain at 6% strain/minute ** ** ____________________ ** ** STEP: Step-1 ** *Step, name=Step-1, nlgeom, amplitude=RAMP, inc=1000 *Soils, consolidation, end=PERIOD 1., 30., , , *El Print, elset=shearstress, FREQUENCY=30 COORD, FLVEL *CONTROLS, PARAMETERS=FIELD, FIELD=PORE FLUID PRESSURE B.8 Submodel for 3% strain at 2% strain/minute ** ** STEP: Step-l ** *Step, nlgeom, inc=1000 *Soils, consolidation, end=PERIOD 1., 130., , , *El Print, elset=shearstress, FREQUENCY=30 COORD, FLVEL *CONTROLS, PARAMETERS=FIELD, FIELD=PORE FLUID PRESSURE name=Step—l, amplitude=RAMP, _--——_--—_-_-—--_-—_ TE u1771111173111:(QIIIIIIMQII