9:337 _UBRARY l Michigan State University This is to certify that the thesis entitled MUSCLE ATROPHY AND ALTERATION OF METABOLIC GENE EXPRESSION DURING EARLY STREPTOZOTOCIN INDUCED DIABETES presented by Erika Templeton has been accepted towards fulfillment of the requirements for the MS. degree in Physiology Wow: Major Professor’s Signature % (2f 1001 Date MSU is an affinnative-action. equal-opportunity employer 0 ’ ' 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 6/07 p'lClRC/DaIeDue.indd-p.1 MUSCLE ATROPHY AND ALTERATION OF METABOLIC GENE EXPRESSION DURING EARLY STREPTOZOTOCIN INDUCED DIABETES By Erika Templeton A THESIS Submitted to Michigan State University in partial fulfillment of the requirements for the degree of MASTER OF SCIENCE Department of Physiology 2007 ABSTRACT MUSCLE ATROPHY AND ALTERATION OF METABOLIC GENE EXPRESSION DURING EARLY STREPTOZOTOCIN INDUCED DIABETES By Erika Templeton Diabetes mellitus can lead to detrimental consequences on organ systems, including that of skeletal muscle degeneration. A model emulating the effects of type I diabetic insulin deficiency can be created in a murine model using the pharmacological administration of streptozotocin (STZ). When treated with acute STZ, mice undergo rapid weight loss and heightened blood glucose. Components of the insulin/IGF-l signaling cascade were examined to determine whether protein degradation and protein synthesis mechanisms were altered. PGC-la, a key regulator of metabolism as well as a putative preventor of muscle atrophy was also investigated. To define the role of STZ diabetes on muscle atrophy and altered expression of metabolic genes, several hindlimb muscles of the diabetic mice were examined in parallel. In STZ mice, weight loss was apparent accompanied by decreased muscle weight in the tibialis anterior. By contrast, the muscle weights of the soleus and plantaris were unchanged. As verification of muscle atrophy, molecular markers of atrophy were examined. MuRFl was unchanged while atrogin-l mRN A expression increased in the plantaris and tibialis anterior. Akt-2 phosphorylation in the tibialis anterior and plantaris were also decreased, corresponding to the increase in atrogin-l. These results indicate that in early acute STZ diabetes, insulin signaling cascade encourages muscle atrophy in the predominantly glycolytic muscles. In contrast, the soleus displayed decreased expression of PGC-l at this time period while the plantaris and tibialis anterior remained unaltered. Thus, different muscles respond differently in onset of muscle atrophy as well as metabolic regulation during the early stages of acute STZ diabetes. Dedicated to my parents, Dave and Lynn Templeton, and brother, Michael Templeton, who continually provide their support in all of my endeavors. iii ACKNOWLEDGEMENTS I would like to thank my committee members, Dr. Robert Wiseman, Dr. Laura McCabe, and Dr. Ron Meyer for their support in this project. I would specifically like to thank Dr. Wiseman for his guidance, patience, and support in the completion of this study. Much thanks goes out to the McCabe laboratory, for providing the animals used in this project and for their continued assistance. I would like to thank Dr. David Ankrapp for his technical support in this project as well as Dr. Arthur Weber and Dr. Thomas Adams for their guidance in completion of this BS/MS degree. iv TABLE OF CONTENTS LIST OF TABLES vi LIST OF FIGURES vii KEY TO ABBREVIATIONS viii SPECIFIC AIMS 1 INTRODUCTION 3 BACKGROUND AND SIGNIFICANCE 5 Diabetes Mellitus ............................................................................................................ 5 Insulin Signaling ............................................................................................................. 7 Mechanisms of Atrophy .................................................................................................. 8 Protein Degradation ................................................................................................... 9 Protein Synthesis ....................................................................................................... 11 Muscle Composition ..................................................................................................... 11 Metabolic Control: PGC-la .......................................................................................... 13 Type I Diabetes Model: Streptozotocin ........................................................................ 15 Current Project .............................................................................................................. 16 SPECIFIC AIM I i 19 Introduction 19 Materials and Methods .................................................................................................. 19 Preliminary Data ........................................................................................................... 26 Preliminary Conclusions ............................................................................................... 35 Continued Research ...................................................................................................... 37 SPECIFIC AIM H 39 Introduction ................................................................................................................... 39 Materials and Methods .................................................................................................. 40 Preliminary Data ........................................................................................................... 43 Preliminary Conclusions ............................................................................................... 48 Continued Research ...................................................................................................... 54 SPECIFIC AIM III - -- 57 Introduction ................................................................................................................... 57 Materials and Methods .................................................................................................. 58 Preliminary Data ........................................................................................................... 59 Preliminary Conclusions ............................................................................................... 61 Continued Research ...................................................................................................... 64 DISCUSSION ’ 67 BIBLIOGRAPHY 72 Table 1. Primer Sequences. LIST OF TABLES vi 25 LIST OF FIGURES Figure 1. Insulin Receptor Signaing Pathway. Figure 2. Mechanisms of Atrophy. Figure 3. Blood Glucose levels. Figure 4. Total Body Weight. Figure 5. Average Muscle Weights. Figure 6. Comparison of cDNA Preparations. Figure 7. Comparison of Housekeeping Genes. Figure 8. Atrogin-l mRNA Expression. Figure 9. MuRFl mRNA Expression. Figure 10. Western Blot Analysis: Akt-2 Figure 11. Western Blot Analysis: IRS-l. Figure 12. Western Blot Analysis: p70 S6 kinase Figure 13. PGC-la mRNA Expression. vii 17 18 28 29 30 31 32 33 34 45 46 47 6O STZ SDS RT-PCR DTT NTP RNA DNA cDNA KEY TO ABBREVIATIONS Streptozotocin Sodium Dodecylsulfate Reverse Trancription-Polymerase Chain Reaction Dithiothreitol Nucleotide triphosphate Ribonucleic acid Deoxyribonucleic acid Complementary Deoxyribonucleic acid viii SPECIFIC AIMS PROJECT Insulin is necessary for the majority of glucose uptake from the bloodstream and is critical for energy homeostasis; however, this hormone is compromised in diabetes mellitus. This disease causes muscle degeneration with a consequent loss in total body weight as well as changes in metabolic regulation. This change in metabolic homeostasis is likely induced by alterations in the insulin Signaling pathway influencing protein expression as well as gene regulation of energetic processes. We propose to test the hypothesis that streptozotocin (STZ) diabetes induces muscle atrophy and altered regulation of metabolic genes. The current study investigates the role of the known insulin signaling pathway in skeletal muscle atrophy using a mouse model of streptozotocin induced diabetes. SPECIFIC AIM I Determine whether early STZ diabetes influences muscle weight and genetic markers of muscle atrophy. Physiologic measurements and quantitative real time PCR will be used. SPECIFIC AIM II Determine whether signaling enzymes that influence protein degradation and synthesis are altered in STZ diabetes. Western blot analysis and animal model measurements will be used. SPECIFIC AIM III Test whether streptozotocin diabetes alters expression of PGC-la, a transcriptional regulator of glucose metabolism and overall cellular metabolic balance. Quantitative real time PCR will be the used. INTRODUCTION Diabetes mellitus poses health threats by producing an abundance of complications in many organ systems that include the cardiovascular, neurological, gastrointestinal and skeletal muscle systems. The purpose of this study is to address the role of streptozotocin (STZ) induced muscle atrophy and metabolic gene regulation during diabetes. In both human diabetic patients and animal models of diabetes, skeletal muscle deterioration and remodeling ensues (Feczko et al. 1994; Oberbach et al. 2006). In uncontrolled diabetes, muscle loss occurs immediately following nutrient imbalance. This evidence suggests the onset of diabetes signals skeletal muscle remodeling very early in the disease. In the absence of insulin, ubiquitin li gases capable of protein degradation are actively transcribed while protein synthesis machinery is also affected (Kandarian et al. 2006). Therefore, determining the mechanism of atrophy requires inspection of both protein synthesis and degradation pathways. Skeletal muscle system is comprised of many different types of proteins and isoforms that offer specialization among each of the muscles. Further complicating the investigation of muscle is the varying composition of muscle types. Following exercise, peroxisome proliferator activated receptor-y coactivator-l (PGC—l), a protein responsible for upregulation of genes involved in oxidative metabolism is highly expressed. Although the increase in PGC-la expression in hypertropht has been well-characterized, the role it plays in atrophy, if at all, is unclear. When PGC-la is overexpressed, muscles are more resistant to atrophy (Sandri et al. 2006). This group also suggests that PGC-la is capable of physically interacting with FOXO, implicating PGC-la as an effector of the insulin signaling cascade. PGC-la levels have been shown to decrease with type H diabetes in muscle (Patti et al. 2003). The role of PGC-la STZ diabetes mediated muscle atrophy is not entirely understood. The goals of the following proposed project are to: 1) demonstrate skeletal muscle atrophy is induced by streptozotocin (STZ) diabetes; 2) elucidate activation levels of insulin receptor signaling components in early type I diabetes; 3) determine activation levels of the insulin dependent degradation pathways; 4) determine activation of the insulin-regulated protein synthesis pathway; 5) ascertain whether PGC-la expression is altered in the muscle during STZ diabetes. Preliminary experiments have been completed for each of the Specific Aims that support proposed experiments. BACKGROUND AND SIGNIFICANCE Diabetes Mellitus Diabetes mellitus is a disease affecting over six percent of the United States population and growing in epidemic proportions according to the Center for Disease Control (Center for Disease Control 2005). Diabetes is the sixth leading cause of death but could be larger because many diabetes related deaths are underreported as a cause (Center for Disease Control 2005). Currently, there is no known cure for diabetes, but successful management can occur through diet, exercise, and pharrnacologic intervention. However, even with treatment, the disease may generate debilitating consequences and an overall poor prognosis. Long term consequences include retinopathy, neuropathy, osteoporosis, kidney disease, stroke, heart disease, and myopathy (Center for Disease Control 2005). Three different forms of diabetes mellitus have been characterized. Though all types are marked by hyperglycemia, they can be distinguished by their origination and progressiveness. Gestational diabetes is acquired during pregnancy in women who do not normally have diabetes (Phillips et al. 2006). Type H diabetes, formerly called adult- onset, is chronic and progressive. Insulin resistance leads to overproduction of insulin itself eventually compromising B-cell function in the pancreatic islets of Langerhaans. Type I diabetes, once known as juvenile, actually occurs with approximately the same frequency in adults and juveniles (National Institutes of Health 2006). Patients with type I diabetes mellitus develop hypoinsulinemia and consequently hyperglycemia due to lack of insulin. Currently, the mechanism of B-cell destruction is unknown, but potential factors are thought to be genetic, autoimmune, or environmental. In all forms of diabetes mellitus, glucose, protein and lipid balance is altered (Beck-Nielson et a1. 1992). The metabolic imbalance produced by diabetes causes altered gene expression and protein modification (Antonetti 1995; Crozier et al. 2003). Alterations at the protein level allow the body to compensate for perturbations in homeostasis; however, these modifications can be maladaptive. With disease progression, further health problems arise as a consequence of diabetes mellitus. One complication attributable to hyperglycemia caused by diabetes is retinopathy, eventually leading to blindness. Eventually, cardiopathy may become a problem coinciding with obesity, hyperlipidemia and hypertension. Diabetes associated neuropathy and osteoporosis also develop (Bouillon et al. 1995). Myopathy is a prolonged consequence of diabetes, but immediate alterations are also visible. In the long term, muscle may begin to be altered in growth patterns (Feczko et al. 1994). In the study by Feczko et al. (1994) mice were induced with type I diabetes by pharmacologic means. After 42 days of diabetes, the extensor digitorum longus muscle of the mouse hindlimb was inspected histochemically. The authors found a shift in the distribution of fibers and destruction of the normal “checkerboard” pattern of fiber distribution (Feczko et a]. 1994). This data displays the impact that STZ diabetes has on muscle over an extended period of time. Skeletal muscle comprises 40-50% of total body mass (Song et al. 1999). It provides structural and mechanical support for the body, but also serves as a potential reservoir of amino acids that can be utilized when necessary. Furthermore, skeletal muscle is responsible for the majority (70%) of total glucose uptake from the bloodstream (Mas et al. 2006). In order for skeletal muscle to accomplish this feat, insulin is a necessary component (Zierath et al. 2000). When insulin binds to receptors on the outer membrane of the muscle cell, it elicits a cascade that ends with the translocation of more glucose transporters to the surface of the muscle in order to relieve the bloodstream of hyperglycemia. As insulin signaling is lost in type I diabetes, the ability to signal the muscle to take up more glucose is also inoperative. Without insulin in appropriate proportions, the muscle perceives that there is not enough available energy in the body. At this point, the muscle begins to breakdown its own supply of proteins in order to accommodate the negative energy balance. This is maladaptive in the diabetic condition since there is an abundance of glucose available for use in the bloodstream, but it is unable to enter the skeletal muscle. When the muscle begins to breakdown more than it is able to build, it is referred to as muscle atrophy or wasting. The signaling components and mechanisms that lead to muscle atrophy have not completely been resolved, though during diabetes lack of insulin may play a role. In this study we investigate the role of insulin signaling using a model that mimics the insulin loss as seen in type I diabetes mellitus. Insulin Signaling The insulin signaling cascade is depicted in Figure 1, initiating with binding of the ligand to the receptor. As a tyrosine kinase receptor it has the ability to autophosphorylate on the B-subunit at multiple residues in order to activate further Signaling proteins (Kahn 1994). Insulin receptor phosphorylation consequently initiates phosphorylation of the IRS (insulin receptor substrate) adaptor proteins thus causing the signal to be passed (Virkamaki A et a1. 1999). IRS is able to activate phosphatidylinositol (PI) 3-kinase which then causes activation of Akt, also known as protein kinase B (PKB) (Oku ,et al. 2001). Akt is activated by phosphorylation at amino acid positions Thr 308 and Ser 473 by phosphatidylinositol 3, 4, 5-triphosphate dependent protein kinase (PDK) (Morino et al. 2006). PI3K and Akt have the ability to transduce Si gnals across multiple pathways imposing regulation on enzymatic processes. This intricate signaling cascade also employs proteins that exist in multiple isoforms. Several isoforms of IRS adaptor proteins have been identified although IRS-l and IRS-2 predominate in skeletal muscle (Bouzakri et al. 2006). Bouzakri et al. showed in myotubes that IRS-2 is dispensable in insulin Signaling while IRS-1 is necessary (2006). Three isoforms of Akt have been shown to exist, but this group determined that Akt—2 is essential to insulin signaling. They concluded that IRS-1 and Akt-2 are crucial to myoblast differentiation as well as glucose metabolism. When the insulin Signaling cascade reaches PI3K and Akt multiple proteins can be activated. The putative scheme of atrophy mechanisms under the control of P13K are displayed in Figure 2. The major pathways associated with insulin Si gnaling include the caspase-3 apoptotic pathway, FOXO transcription factors, and mTOR (mammalian target of raparnycin) pathway. Activation of FOXO and mTOR are mediated through Akt Signaling. These pathways are discussed in the following section. Mechanisms of Atrophy Skeletal muscle responds quickly to drastic declines in energy demands, rapidly degenerating proteins into amino acids that can be used for energy. During diabetes, muscle continually remodels. When the muscle is in a state in which protein degeneration exceeds protein synthesis, it is referred to as atrophy. Atrophy has been characterized by Lecker and colleagues (2004) as both a decrease in protein synthesis and an increase in protein degradation. Protein Degradation The first Akt-mediated mechanism of muscle breakdown in need of investigation involves members of the ubiquitin proteosome pathway. The drastic increase in muscle proteolysis is due to an increase in ubiquitination (Lecker et al. 2004). Degradation via the ubiquitin proteosome path is ATP-dependent and involves li gases that tag proteins destined for degradation with a string of ubiquitin molecules (Hershko et al. 1998). Ubiquitination allows the proteins to be identified and destroyed by the 26S proteosome (Hershko et al. 1998). Many ubiquitin ligases are thought to exist, but two are dramatically upregulated during Skeletal muscle atrophy. Atrogin-l/MAFbx (muscle atrophy F box) was originally identified simultaneously by Gomes et al. (2001) and Bodine et al. (2001). Discovery of MuRFl was also accomplished by Bodine and colleagues (2001). Both atrogin-l and MuRF 1 have been shown to increase in thirteen different forms of atrophy (Bodine et al. 2001). Collectively atrogin-l and MuRFl can be referred to as atrogenes. Found in skeletal and cardiac muscle, atrogin-l and MuRFl have been characterized as E3 ubiquitin li gases (Cao et al. 2005). The Ub-protein ligases are specific for a certain class of proteins to mark for breakdown (Hershko et al. 1998). The E3 portion of the enzyme is possesses a recognition site to confer specificity on the substrate (Passmore et al. 2004). Several groups have demonstrated that atrogin-l plays a role in both initiation and overall maintenance of protein degradation (Gomes et al. 2001; Lee et al. 2004; Sandri et al. 2004). Both atrogin-l and MuRFl null mice appear phenotypically identical to litterrnates until they undergo hindlimb suspension, whereby the null mice are resistant to muscle atrophy. The atrogin-l null mouse exhibits a greater resistance to muscle wasting than the MuRFl null mouse model (Bodine et al. 2001). Lecker et al. Showed that in many forms of skeletal muscle atrophy, atrogin~1 and MuRFl were upregulated in mRNA expression (2004). As these genes are shown to readily increase with atrophy, they can be used as molecular markers of muscle wasting. Atrogin-l expression is controlled through Akt signaling. Transcription of this ubiquitin ligase is controlled directly by the FOXO family of transcription factors (Lee et al. 2004; Sandri et a1. 2004). FOXO transcription factors are effective activators of atrogin-l as FOXO is able to bind directly to the promoter for atrogin-l (Sandri et al. 2004). Though the target of atrogin-l is not completely clear, MyoD and calcineurin have been identified as potential substrates (Lee et al. 2004; Tintignac et al. 2005). MuRF-l (muscle-specific RING finger protein 1) is a 40 kDa protein belonging to the family of TRIM proteins or tripartite because it contains three important domains (Bodine et al. 2001; Kamura et al. 1999; Centner et al. 2001). These domains include a RING finger domain, allowing for ligase activity, a B-Box domain with an unknown function, and a coiled-coil domain which may be necessary for the formation of heterodimers (Kamura et al. 1999; Centner et al. 2001). Kedar et al. (2004) have shown that MuRFl has the ability to bind cardiac troponin I, while Weinert et al. (2006) determined that the structural protein titin contains a binding site for MuRF 1. FOXO transcriptional activators regulate atrogin-l expression, but whether they are able to activate MuRFl is controversial. When phosphorylated, FOXO is contained by 14-3-3 proteins in the cytosol (Nader 2005). However, upon dephosphorylation they 10 readily translocate to the nucleus to influence transcription (Sandri et al. 2004). To date, three mammalian isoforms have been identified (Nader 2005). These forms include FOXOl (FKHR), FOXOBa (FKI-IRLl) and FOXO4, all of which are thought to be under the dominant control of Akt (Nader 2005). Protein Synthesis As well as activating means of protein degradation, Akt also controls protein synthesis. Phosphorylated Akt facilitates the phosphorylation of mTOR (mammalian target of rapamycin) kinase. This protein has the ability to cause phosphorylation of p70 S6 kinase, which as its name suggests, is able to phosphorylate the p70 subunit of the S6 ribosome (Fingar et al. 2004). S6 ribosome phosphorylation allows protein production to increase. mTOR is also able to phosphorylate 4E-BP1 (Elongation factor 4E binding protein 1), which relieves suppression on 4E, encouraging translation (Kandarian et al. 2006). Overall the mTOR pathway is highly active when nutrients are present allowing muscle hypertrophy to occur (J ackman et al. 2004). Conversely, inactivation of this pathway leads to decreased protein synthesis, resulting in atrophy. Studies have Shown that FOXO and mTOR pathways are regulated by Akt signaling. As a member of the insulin receptor signaling cascade, Akt activation may be altered by diabetes. Muscle atrophy occurs during uncontrolled diabetes and at different rates depending on the muscle type used (Lecker et al. 2004; Thomason et al. 1987). The complete mechanism as to how atrophy occurs is unclear, while the process that makes atrophy occur at different rates in muscles is particularly vague. Muscle Composition ll Thomason and colleagues (1987) showed that wasting occurs at different in various muscles; however, it is unclear why this is the case. One explanation may be substrate preference between the muscle fiber types. Another reason could be that the functional load between muscles varies so that the muscles can selectively remodel according to necessity. A third explanation may be that muscle contains different concentrations of protective proteins which differ between fiber type. Though the difference in atrophy rates is unclear, the variation between muscle types has been characterized. To fully grasp how muscle atrophy may occur at different rates an understanding of the muscle composition by type in necessary. Muscles are categorized into three predominant fiber types based on several distinguishing factors. Fibers can be categorized according to myosin ATPase isoform. As a contractile protein, myosin is a major contributor to differences in speed and sustainability of contraction. According to their method of energy production, muscles are classified as oxidative or glycolytic. Oxidative fibers or slow twitch, have increased ability to perform oxidative respiration, fatty acid oxidation, increased triglyceride storage, and a lower glycolytic capacity in comparison to glycolytic muscle fibers (Nemeth et al. 1979). In order to have increased levels of oxidative respiration, type I fibers contain greater amounts of mitochondria. Furthermore, the fiber types contain different sets of enzymes and protein isoforms (Puigserver 2005). Glycolytic muscles rely mainly on glycolysis for their energetic demands and cannot sustain contraction for extensive time periods. Glycolytic muscle fibers are further divided into type Ila (oxidative glycolytic) and type IIb (fast glycolytic) fibers. Type IIa fibers have a greater oxidative capacity and are more fatigue resistant 12 than type IIb glycolytic fibers (Saltin et al. 1983). Variations in muscle fiber proteins provide the necessary functional capacity. In experimental diabetes, histochemical analysis of muscles shows that fiber type switching may begin to occur from a Slow to fast phenotype (Feczko et' al. 1994). Niederle et al. (1978) demonstrated a Shift from type 2A to 2B in denervated rats. It was also Shown that 2B fibers underwent atrophy more quickly than 2A fibers (Niederle et al. 1978). In time course study of hindlimb unweighing, Thomason et al. Showed that the rate and degree of muscle loss differed according to muscle type (1987). In their model, the soleus muscle was the most rapid to undergo atrophy (Thomason et al. 1987). Furthermore, phosphorous containing metabolites and regulatory enzyme functions are also altered in a fiber-specific manner during diabetes (Fewell et al. 1995). Although skeletal muscles have distinct properties for functionality, all are void of insulin signaling during type I diabetes. Despite substrate selectivity and load of the muscle, different fibers have variations in gene activation. The reason for the disparity in degeneration of muscles is unknown. Metabolic Control: PGC-la A primary difference between muscle types is the proportion of glycolytic and oxidative fibers composing the muscle. The transcriptional coactivator PGC-la is capable of controlling transcription of metabolic genes. In type I diabetes, loss of insulin signaling causing metabolic imbalance in the muscle, therefore PGC-la activity may be altered. PGC-la may also influence FOXO and Akt activity, two key components in muscle atrophy (Sandri et al. 2006). Much research has characterized the role of PGC-la 13 expression in response to exercise, but less has been revealed as to how PGC-la expression is influenced by type I diabetes. Metabolic pathways achieve control partially at the level of gene expression. Transcription factors exert a level of control on gene expression, but coacti vators are able to modulate the activity of the transcription factors. In skeletal muscle, PGC-la has been shown to have an important role in mitochondrial biogenesis and fiber type switching. Fiber type switching causes muscle fibers to change phenotypes, effectively altering the exercise capacity of the muscle. Sandri and colleagues (2006) took many forms of atrophy into account including cancer, cachexia, diabetes, renal failure, and denervation in order to determine that PGC- la mRN A levels decreased among the tibialis anterior muscles that were atrophic (Sandri et al. 2006). This group also created transgenic mice overexpressing PGC-la in Skeletal muscle. They subjected these mice to denervation and fasting, discovering that the transgenic mice had less muscle atrophy and decreased induction of atrogenes than the control mice. They concluded that the ability of FoxO3 to bind at the atrogin-l promoter was decreased in the PGC-la overexpressing mice (Sandri et al. 2006). This group suggested that PGC-la physically inhibited the actions of FoxO3 preventing the induction of atrophy. FOXO controls expression of atrogenes within the insulin signaling cascade, further implicating PGC-loi alteration in diabetes and muscle atrophy. Other studies have shown that overexpression of PGC-loi increases mitochondria and myoglobin content, key components of slow muscle fibers (Lin et al. 2002). Furthermore, PGC-la is preferentially expressed in type I muscles (Puigserver 2005). 14 PGC-la clearly has a role in metabolism and a connection to the FOXO family, though these roles during muscle atrophy are in need of clarification. Type I Diabetes Model: Streptozotocin Multiple forms of animal models have been developed to emulate the diabetes mellitus. Genetic and pharmacologic models both exist for further manipulation and study. One pharmacologic mouse model of type I diabetes mellitus can be created in the lab using the drug, streptozotocin (Like et a1. 1976). The chemical structure of streptozotocin is similar to a glucose molecule enabling it to enter cells bearing the GLUT2 glucose transporter. GLUT2 glucose transporters are found in the hypothalamus, basolateral side of the small intestine, liver and in a larger proportion on the B-cells of the pancreatic islets. Other glucose transporters are located in other tissues; however, STZ is unable to enter. As the drug enters the B-cells, it elicits a cytotoxic effect by causing damage to the DNA by alkylation (Szkudelski 2001). Using multiple low doses of STZ, the development of diabetes can be imitated (Like et al. 1976). When diabetes is induced by this method, the animals Show decreased growth, hyperglycemia, hypoinsulinerrria, and hyperlipidemia (Chen et al. 1982). Acting as a cytotoxin, the question whether STZ has effects on other cell other than the B-cells of the pancreas may arise. All tissues rely on glucose uptake for survival, which hold a potential role for STZ to elicit a negative effect. When comparing the histology of STZ to genetic models of diabetes, Skeletal muscle morphology appears very similar (Feczko et al. 1994). This would suggest that the drug does not have a negative effect directly on the muscle and that the alterations visible in muscle are a consequence of the metabolic imbalance created by streptozotocin. 15 Current Project The purpose of this current project is to determine the actions of diabetes on Skeletal muscle. Understanding the influence of diabetes on skeletal muscle will help to understand the impact of diabetes in other organ systems. Diabetic models that undergo rapid muscle degeneration are useful to provide insight into the dynamic nature of skeletal muscle and their overall role in total body metabolic balance. This research has investigated a portion of this problem while suggesting further projects to continue this inquiry. At this point, this research begins to identify a portion of the body’s mechanism in response to early STZ diabetes. This project examined the insulin signaling components that control protein catabolism and anabolism without consideration of converging pathways so that these specific paths may be isolated. Also considered was PGC-la, an important component to metabolic control and potential mediator of muscle atrophy. The diabetic model here was examined two days following the final acute injection of streptozotocin. Along with preliminary data presented, further experiments are proposed that would investigate a pathway that may explain the difference in atrophy between muscles. Additional experiments will allow Speculation about what cellular components are important in the prevention of muscle wasting. Although research has characterized the effect of Streptozotocin in Skeletal muscle, few studies have examined different muscle types in parallel and at any early time of STZ treatment. This project may help to elucidate the role of diabetes in early muscle atrophy in differing muscle types, which will assist in directing further research on atrophy in skeletal muscle. 16 Insulin Insulin L Receptor "' ' \\ . (f . IRS-1 I p85 PISK W .1 + ..5KT_.. / \ mTOR FOXO (protein synthesis) (gene expression) Figure 1. Insulin Receptor Signaling Pathway. This diagram depicts some of the proteins affected by the binding of insulin to the insulin receptor. When insulin binds, tyrosine autophosphorylation of the receptor occurs resulting in the phosphorylation of IRS-1 (Insulin receptor substrate-1). This causes phosphorylation of PI3K (phosphatidyl inositol-3 kinase) which is able to influence phosphorylation of Akt. Akt is then able to influence protein synthesis through the mTOR pathway and gene expression through a FOXO-mediated path. This diagram has been modified from Kido et al. (2001). 17 Deactivation/ dephosphorylation ® ic-Pisk / 4E-BP1 *6.) f activation , , T C -3 deactivation Decreased Increased 339339 (9 ix- AKT —' mTOR *6“) protein . I I synthesis apopoSIs Aw" fragmentation p70 36K *6) J Ub proteosome! degradation Cytoplasm uni 't' it Nucleus r / 0334,17,? (D It FOXO activation AII'O in-1 Increased L , /' g Proteolysis AITOQ'N'I L Activation of other atrophy genes (MuRF1) Figure 2. Mechanisms of Atrophy. Putative atrophy signaling pathways associated with diabetes mellitus. Boxes indicate proteins, phosphorylation is denoted by circles. With dephosphorylation of PI3K, its activity is decreased. This causes activation of caspase-3 and deactivation/dephosphorylation of Akt. Akt is able to influence both catabolic and anabolic pathways. With dephosphorylation of F OX0, atrogin-l is actively transcribed. When Akt causes dephosphorylation of mTOR, protein synthesis is decreased by dephosphosphorylation of 4E-BP1 and p70 S6 kinase. Diagram has been modified from Kandarian et al. (2006). 18 SPECIFIC AIM I INTRODUCTION The first question that must be considered is whether the STZ diabetic model is an appropriate model for the determination of early skeletal muscle wasting. In order to destroy the B cells of the pancreas to ‘prevent insulin production, STZ was administered. This causes the mouse model to become comparable to type I diabetes mellitus. Other studies have shown that with administration of STZ, hyperglycemia, hypoinsulinemia, and rapid weight loss occur in the animal (Cheng et al. 1982). Despite a suspected decrease in total body weight with STZ, it needs to be confirmed whether this model is losing weight in the form of skeletal muscle. In multiple causes of atrophy, data has Shown two ubiquitin ligase mRN A levels to be increased specifically in muscle (Bodine et al. 2001). Atrogin-l and MuRFl can therefore be used as markers of skeletal muscle atrophy. These genes are capable of increasing in expression before measurable weight loss occurs (Bodine et al. 2001; Gomes et al. 2001). This report will focus on the physiologic data presented by the model of diabetes as well as the genetic markers of atrophy. Data could then be expanded upon to determine the methods of achieving consistent results with experimental STZ diabetic animals helping to understand the impact of diabetes on muscle in more complex models. Specific Aim I investigates the modification by STZ diabetes on body weight, muscle weight and activity of atrophy markers in the skeletal muscle. MATERIALS AND METHODS 19 Animals Adult male (15 week old) BALB/c mice (Harlem Laboratories, Houston, TX) were kept in 12 hour light-dark cycle at a temperature of 23 C and had access to food (standard lab chow) and water ad libitum. All mice were housed in the Michigan State University Biochemistry Animal Facility. Animal studies were performed in agreement with the Michigan State University All-University Committee on Animal Care and Use. The animals were examined daily. Diabetes was induced using intraperitonial injection of streptozotocin (40 ug/ g body weight in 0.1 M citrate buffer), a pancreatic beta cell cytotoxin, over the course of five days (Like et al. 1976). The injections occurred during the same time period each day. Control mice were injected with buffer alone and the treated and control mice were kept in separate cages with 3 to 4 mice per cage. The tissues were harvested approximately 24 hours following the last injection for the mice sacrificed days 1 through 5. The animals were euthanized using a carbon dioxide chamber with the hindlimb immediately removed following sacrifice. We removed the tibialis anterior, plantaris and soleus muscles from one of the hindlimb. Blood glucose levels were measured post- mortem using a glucometer (Accu-Check instant, Roche Molecular Biochemicals Corp., Indianapolis, IN). The muscles were immediately frozen in liquid nitrogen and stored at - 80 °C until further use. Muscle weights were taken prior to running an assay. Experimental Groups The mouse experimental groups were divided into eight groups to be sacrificed 1, 3, 5 and 7 days following initial injection of streptozotocin, each group containing control 20 and diabetic mice. Six control mice and eight treated mice composed each of the groups. The focus of this project was the seven day group Since this would allow the mice two days to recover from the last injection. These two days could allow any stress response to the injection protocol to subside in the mice. Choosing this time point also provided time for the pancreatic B cells to fully undergo cell death (Pi ghin et al. 2005). Furthermore, at this time point the animals reached a heightened blood glucose level of 300 mg/dl, varying greatly from the control. Given the small Size of the muscles and multiple assays to be run on each muscle sample, a number between 5-6 mice were used for the controls and between 7-8 mice for the diabetic mice tissues. Tissue Preparation In order to utilize the muscles for multiple assays, the muscle was ground or halved under liquid nitrogen and distributed into multiple vials. This was accomplished by pestle and a mortar filled with liquid nitrogen and performed in a cold room. Prior to pulverization or fractionation, the muscles were weighed while frozen and the weight was recorded. The weight of each tissue distributed to each vial was also measured and recorded. Different fiber types are distributed throughout the tibialis anterior so in order to distribute the tissue more uniformly in each vial, it was necessary to crush the tissue. The soleus and plantaris have more uniform fiber type distribution so could simply be halved in order to use the tissues for separate assays. Also, as these muscles are much smaller than the tibialis anterior, it was important to conserve as much tissue as possible. The tissue was stored at -80 °C until it was used for further analysis. Reverse Transcription and Quantitative Real Time PCR 21 The total RNA was isolated from the mouse muscles using Trizol reagent (Invitrogen). The tissues were homogenized in the reagent then placed in a 37° C water bath to allow for dissociation of nucleoprotein from the nucleic acids. Chloroform and glycogen were added, the homogenate was placed in the centrifuge and following centrifugation, the supernatant containing RNA was removed and isopropanol was added. The RNA containing mixture was placed at 4° C overnight to allow for precipitation of RNA to occur. The RNA precipitate was washed in ethanol and stored in forrnarnide at - 70° C for further use. Reverse transcription of the RNA to cDNA was accomplished using a mixture of Superscript H (Invitrogen), dNTPs, dithiothreitol, DEPC-water, RNAse inhibitor (Invitrogen), 5x First Strand Buffer (Invitrogen) and primers (Invitrogen) in a 20 til total reaction volume. A maximum of four primers were mixed in the primer mix at a time and sequences were compared to discourage interaction of the primers. The mixture was incubated at 42 °C for 60 minutes, followed by 72 °C for 15 minutes. In order to confirm that the cDNA preparations were not varying greatly, separate cDNA samples were prepared for some tissues. These separate cDN A samples were then analyzed for gene expression using quantitative RT-PCR. Quantitative real time PCR was carried out using the iQ Sybr Green Supermix System, the iCycler and iCycler software for analysis (BioRad). Experiments were performed in duplicate with the following parameters: rrrinutes at 95 °C for 3 minutes followed by 40 cycles of 95 °C for 30 seconds, 58 °C for 60 seconds and finally with 72 °C for 30 seconds. To ensure the quality of analysis, a negative control (sample replaced 22 with RNAse free water) and a positive control consisting of a pool of positive samples was used each time the qRT-PCR was run. B-actin was used as the housekeeping gene and results were expressed using the comparative threshold cycle of B-actin and the gene of interest. In order to confirm that B—actin mRN A was acting as an appropriate control, some samples were analyzed for cyclophilin as well as B-actin. B-actin is a ubiquitous cytoskeletal protein and has been Shown to be stably expressed following resistance exercise in skeletal muscle (Mahoney et al. 2004). Cyclophilin is found in the cytosol and functions in cellular protein folding and interactions. The samples that were chosen for comparison of the housekeeping genes consisted of both the plantaris and tibialis anterior as well as control and diabetic muscles. The samples were run from the same cDNA preparations and were run through quantitative real time PCR at the same time as well to ensure conditions were identical. Therefore, any differing results could be attributable to the housekeeping gene. Furthermore, expression levels of two target genes were compared to further assess the reference genes. All primers were verified using BLAST which determined that the sequence homology was specific for the target gene. The primers were purchased from Invitrogen. Forward and reverse sequences for each primer can be found in Table 1. Threshold Cycle (CT) values for the Specific and control genes were obtained automatically by the BioRad software. The data was then analyzed using the 2 ‘AACT method (Livak et al. 2001). The relative level of the specific gene of interest (PGC-la, MuRFl or atrogin-l) was determined by substracting the CT values of the housekeeping gene (B-actin or cyclophilin) from the CT value of the gene of interest. 23 Statistical Analysis Statistical analyses throughout this project were performed using Microsoft Excel data analysis program for Student’s t test analysis. The values are expressed as means 1 SE (standard error of the mean). A p value less than 0.05 was considered to be significant. 24 Primer Sequences Atrogin- l/MAFbx Forward 5 ’ -GCAGAGAGTCGGCAAGTC-3’ Reverse 5 ’ —CAGGTCGGTGATCGTGAG-3 ’ MuRF—l Forward 5’-TGTCTGGAGGTCGI I I CCG-3’ Reverse 5 ’ -CAGGTCGGTGATCGTGAG-3 ’ PGC-la Forward 5’ -GCCGTGTGAI I I ACGTTGG-3’ Reverse 5 ’ -GATAGTACACGCTGTCCCATG-3’ B-actin Forward 5 ’ -GTATGCCT CT GGTCGTACCAC-3’ Reverse 5 ’ -CTAGAAGCACTTGCGGTGCA—3 ’ Table 1. Primer Sequences. Forward and reverse primer sequences used to determine mRNA expression with quantitative real time PCR. 25 PRELIMINARY DATA Blood Glucose At day seven, the diabetic mice reached significantly elevated blood glucose levels in comparison to the control mice (Figure 3). The treated mice had a blood glucose level reaching approximately 300 mg/dL, while the control mice were below 200 mg/dL (McCabe et al., unpublished observation). This data corresponds to blood glucose levels found in the literature for mice injected acutely with streptozotocin. Body Weight Within a time span of 7 days following initiation of drug treatment, the body weights of the treated mice were less than the control mice, reaching a statistically significant level. Total body weight decreased approximately 10% in the diabetic mice by day 7. The body weights of the mice for the length of the time course can be found graphically in Figure 4. Muscle Weights Whether there was a difference in muscle weights varied according to muscle (Figure 4). The tibialis anterior experienced approximately a 14% decrease in weight in comparison to the control weight. This weight difference was Significantly different between diabetic and control. The soleus and plantaris muscles showed no apparent difference in muscle weight across the entire time course. cDNA Preparation analysis Separate cDNA samples were prepared in order to determine variability in mRNA analysis. The samples that were repeated showed consistency between the trials. This is depicted in Figure 6. 26 Housekeeping Gene Analysis The use of cyclophilin confirmed the results obtained from using B-actin as the housekeeping gene. In Figure 7, the values are expressed as a percent of the overall average of the expression level. The expression levels of most of the samples corresponded between B-actin and cyclophilin when examining both at i'ogin-l and PGC- la genes. Molecular Markers of Atrophy FOXO activation was assessed by detemrining mRNA levels of atrogin-l and MuRF 1. This data is depicted graphically in Figure 8. The mRNA expression levels in comparison to B-actin were assessed individually for each sample then averaged for the group. In the tibialis anterior, the mRNA expression level of atrogin-l, but not MuRFl was increased in comparison to the control muscles. The increase in expression reached statistical Si gnificance among the atrogin-l mRNA. This suggests that atrophic mechanisms had been induced in this muscle. The plantaris muscle trended toward an elevated level in atrogin-l mRNA expression among the diabetic mice,‘ without obtaining Si gnificance. Again, the MuRFl levels were unaltered between control and diabetic plantaris muscles. In the soleus muscle, the levels of both atrogin-l and MuRFl expression were not significantly altered overall at day 7. The expression levels are compared to the levels of the housekeeping gene, B-actin. In order to assure that B -actin levels were not changing with drug treatment or atrophy, cyclophilin was also assessed as a housekeeping gene for some of the samples. Figure 7 displays validation of B-actin as a housekeeping gene in comparison to cyclophilin. 27 Blood Glucose Levels 350.0 300.0 j , 250.0 - i. 200.0 - I +Control 150.0 ~ i {-l— Diabetic 100.0 . 50.0 ~ 0.0 Blood Glucose (mgldL) \I 1 3 5 Time (days) Figure 3. Blood Glucose Levels. Blood glucose was measured immediately prior to sacrifice. The blood glucose levels reached statistically different values at day 3. At day 7, the blood glucose levels reach 300 mg/dL. (McCabe et al., unpublished observations). (as) Signifies significance. 28 Body Welght 30.0 * 25.0 - Mi! 20.0 . —o— Ctrl 15.0 ~ -—I—— STZ BOW Weight (9) 10.0 « 5.0 J 0.0 Time (Days) Figure 4. Total body Weight. The mice were weighed prior to sacrifice. Each group is representative of the average of 6 control mice and 8 diabetic mice. At day 7, the two groups reach statistically different body weights. (McCabe et al., unpublished observations) 29 Average Muscle Weights 0.06 0.05 - 0.04 — 0.03 ~ 1 Control LI Diabetic: Weight (grams) 0.02 - 0.01 4 Tibialis Anterior Plantaris Soleus Figure 5. Average Muscle Weight. The weights included are not tendon-free. The control and diabetic samples of the soleus and plantaris muscles were not statistically Significant. The tibialis anterior reached a statistically different weight by day 7 between the two groups. (t) signifies significant difference (ps0.05) 30 Comparison of Separate cDNA Preparations _L ix) .4 i 9 on J_ E First Prep I Second Prep ‘ Atrogin-I mRNA expression (A U) C O) 0.4 « 0.2 0 J 1 2 3 4 5 6 7 Samples Figure 6. Comparison of cDNA Preparations. Expression levels of mRNA prepared in separate cDNA reactions. The same RNA mixture was used to prepare separate cDNA samples. The samples were then run in the same quantitative real time PCR reactions. Both atrogin-l and B-actin were analyzed separately for each sample. Both diabetic and control muscles were used. Arbitrary units (A U) 31 Comparison of Housekeeping Genes using Atrogin-I as Target Gene 01 O I .— 250 .é or 2 200 ~ 5 s E’ 3 150 « B-Actin o o .. 3, E 100 1 ICyclophilin ‘5 I.IJ E § 0 n. O M 1 2 3 4 5 6 7 Samples Comparison of Housekeeping Genes using PGC-1 as target gene _A CD 0 160 - _‘ A _L co 0 N Ii 0 O O O l l l B-Actin I Cyclophilin 60* 40‘ 20~ 1" 0— Percent of Average PGC-1 Expressmn 1 2 3 4 5 6 7 8 Samples Figure 7. Comparison of Housekeeping Genes. Atrogin-l and PGC-la were both used as target genes. This graph seeks to validate the use of B-actin as a housekeeping gene in comparison to cyclophilin. Both control and diabetic as well as various muscle samples were utilized. The sample numbers on the two graphs do not correspond to each other. 32 Atrogin-1 mRNA Expression b I I Control I Diabetic AtrogIn-t expresslon (A U) N (a) Tibialis Anterior Plantaris Soleus Figure 8. Atrogin-l mRNA Expression. There was a significant difference among the expression levels of atrogin-l in the tibialis anterior muscle. Although the plantaris trended toward an increase in expression, the difference was not statistically significant. Arbitrary Units (A U) (*) signifies significant difference (p50.05) 33 MuRF1 mRNA Expression 2.5 ‘ A 2 D S, C .3 1.5 — 3 IControl a IDiabetic K O 1 I E I: :I E 0.5 0 - Tibialis Anterior Plantaris Soleus Figure 9. MuRFl mRN A Expression. There was no significant difference between the control and diabetic expression levels for this gene among all three muscles. Arbitrary units (A U) 34 PRELIMINARY CONCLUSIONS Blood glucose levels were significantly elevated in the diabetic mice in comparison to the control (McCabe et al., unpublished observations). Since heightened blood glucose levels are characteristic of diabetes mellitus, it would indicate that these mice are diabetic. Furthermore, the streptozotocin protocol used in this experiment has been utilized frequently as a model of type I diabetes. Since the seven day time point was chosen, it enabled the drug to be given for all of the proposed days, so that destruction of the pancreatic B cells could be achieved fully. Since use of this method before has confirm loss of insulin production, the insulin measurements were not assessed in this study. Body weights indicate a shift towards a decrease in weight, though not to the extent that was anticipated. In unpublished data from the McCabe laboratory, extensive weight loss was visible within a shorter time period when using the $31116 protocol (McCabe et al. 2006). This could be attributable to the demeanor of the handler towards the mice. Nonetheless, approximately 6% of total body weight was decreased in 7 days (Figure 2). The weight loss exhibited by this model confirms results of weight loss following STZ induced diabetes found in the literature. In this study, only one of the muscle displayed a decrease in weight following treatment. The tibialis anterior experienced visible weight loss. The plantaris trended toward a weight decrease while the soleus showed no Sign of weight change. Weight measurements were not tendon-free, which could have created an error in muscle weights. Due to the time constraint during the muscle harvest, the tendons could not be removed. To do so would have compromised the integrity of the tissues. 35 Atrogin-l was induced transcriptionally, indicating that atrophy is or is about to take place. It is interesting that MuRFl expression was unaltered. Bodine et al. (2001) as well as Gomes et el. (2001) determined that these atrophy markers were increased prior to loss in body weight in multiple models of muscle wasting. The atrogin-l data acquired in our study corresponds to their findings. This data may indicate that MuRFl is not under regulation of the FOXO family of transcription factors as atrogin-l has been Shown to be. This explanation is supported by Kamei and colleagues (2004) who showed that overexpression of FoxOl caused atrogin-l expression to rise without a concomitant increase in MuRFl expression. Alternatively, MuRFl upregulation may be induced at a later time period than atrogin-l. Activation of atrogin—l expression corresponded to the decrease in individual muscle weights exhibited by the muscles, suggesting that upregulation of this ubiquitin ligase is occurring. Since atrogin-l is under the dominant control of FOXO, these results suggest that FOXO has been dephosphorylated and translocated to the nucleus in order to influence transcription. According to both muscle weights and activation of atrogin-l, the results were not consistent across the three muscles. The preliminary data suggests that the plantaris may be in the beginning stages of atrophy. Since the tibialis anterior and the plantaris are mainly comprised of glycolytic fibers, it is interesting that they were the first to trend towards muscle atrophy. The data exhibited here point towards a protective role of oxidative muscle during atrophy. Further analysis of the Specific mechanisms causing delay in atrophy of the soleus needs to be assessed. Also in need of consideration is the anatomical location of the muscle, which provides differential stimulation patterns that may contribute to varied results among 36 muscles. The tibialis anterior displayed the greatest increase in activation of atrogin-l, whereas the soleus and plantaris did not show a significant increase. Both of the former muscles are located in the posterior compartment of the mouse hindlimb whereas the tibialis anterior is located in the anterior position. This could indicate that there is a selective loss of muscle fiber during diabetic muscle atrophy according to placement of the muscle in the mouse hindlimb, which may be dependent upon usage. The conclusion, based on preliminary data, is that this model does cause skeletal muscle atrophy within one week of beginning STZ treatment. With this model, atrophy occurs in different muscles at different rates. An understanding of the mechanisms affected by STZ diabetes will facilitate further understanding of muscle atrophy. CONTINUED RESEARCH The mechanism leading to atrophy during diabetes has not been fully elucidated. This study showed that the glucose levels were gradually increasing in the bloodstream of the treated mice. Other research has shown that with administration of STZ diabetes in conjunction with amelioration of hyperglycemia, considerable weight loss still occurs (Cheng et a1. 2006). Further research is necessary to fully characterize the role of glucose and insulin in atrophy. Blood levels of insulin should also be determined in continued research. This could elucidate loss of insulin as a director of muscle atrophy at this time point. It is also interesting that when the same procedure was followed previously, the mouse weight did not decrease to such an extent (McCabe 2006). Continued research could include reproducing the animal trials multiple times. More tissue allotted for 37 mRNA would be beneficial to indicating the levels of the target gene. This would require the use of many more animals. Finally, animals could be analyzed at an earlier time point to determine when atrogin-l is first activated in accordance with the loss of insulin Signaling. It would be expected that further animal trials would supply more data showing earlier activation of atrogin-l prior to a decrease in body weight. Further time points could also allow insight into transcriptional upregulation of MuRFl in different muscle types. One way to test whether hyperglycemia is at fault would be to treat animals as in the STZ protocol with metformin to ameliorate hyperglycemia. Although metformin studies with streptozotocin thus far have revealed that the animals still experience weight loss, it has not been examined at such an early time point as the 7 day period (Suwa et al. 2006). This research would be able to tell us whether hyperglycemia impacts the insulin signaling pathway components directly or if hyperglycemia is the primary reason for Skeletal muscle atrophy in the early stages of atrophy. 38 SPECIFIC AIM IIError! Bookmark not defined. INTRODUCTION When protein degradation exceeds protein synthesis, muscle wasting occurs. Animal measurements and molecular markers of atrophy provided clues that STZ diabetes is an activating response of muscle degradation. Since insulin depletion has been demonstrated to occur in this model, components of the insulin signaling pathway that can influence muscle wasting need to be examined. Western blot analysis is a technique that allows for the differentiation between phosphorylation levels of proteins involved in the insulin Signaling pathway. Si gnaled through Akt, there are two major pathways that contribute to protein synthesis and degradation. These pathways include FOXO and mTOR. Downregulation of Akt causes dephosphorylation and activation of FOXO. Transcriptional upregulation of atrogin-l occurs when FOXO transcription factors are active in the nucleus of the cell. Atrogin-l is able to cause ubiquitination of proteins, tagging them for degradation. Regulation of MuRFl is controversial, but it may also be upregulated by the FOXO family of transcriptional activators (Kandarian et al. 2006). Atrogin-l and MuRFl were investigated in Specific Aim 1. Collectively, these factors promoting muscle degradation are upregulated in many forms of atrophy, however, their activation in early models of streptozotocin induced diabetes have not been completely explored. Also signaled through Akt, mTOR influences rates of protein synthesis through p70 S6 kinase. Decreased phosphorylation of Akt consequently causes a decrease in the phosphorylation levels of mTOR. P70 S6 kinase causes phosphorylation and activation 39 of the S6 ribosome increasing protein synthesis. Current research puts a heavier weight on increased protein degradation over decreased protein synthesis during muscle wasting. Specific Aim II will focus on key components of the insulin signaling cascade during STZ diabetes. Components dealing with protein synthesis and degradation will be investigated in this portion of the project. Although signaling pathways are able to interact, it is important to isolate components for inspection, yet consider alternative influences. Furthermore, oxidative and glycolytic muscles will once again be examined in order to provide insight into signaling mechanisms of different muscles. MATERIALS AND METHODS Animals Animals used were the same as utilized in the Materials and Methods of Specific Aim I. Tissue Preparation The muscles were prepared as in Specific Aim 1. The same tissues were used for comparison in this section of the study. SDS-polyacrylamide gel electrophoresis The tibialis anterior, soleus and plantaris muscles were homogenized in a solution containing 10X TBS, glycerol, 0.1 M dithiothreitol, 0.5 M sodium fluoride, 10 mM sodium orthovanadate, 20 mM sodium pyrophosphate, 0.25 M beta-glycerophosphate and double-deionized water. 20% w/v sodium dodecyl sulfate was added to the homogenate allowing cell lysis to occur. After centrifugation, the supernatant was acquired and subjected to electrophoresis. 40 For all of the protein preparations, an 8% Tris-glycine SDS-polyacrylamide gel were used. The gel sizes were determined according to the molecular weight of the protein. IRS-1 has a molecular weight of 180 kDa, Akt-2 is 60 kDa, p70S6K is 70 kDa and the full version of caspase-3 is 35 kDa in weight. The percentages of gels optimized band separation among the proteins, while allowing the membrane to be used for multiple Western blots. The gel was poured to a thickness of 1.5mm and 10 wells were created within each of the gels. A 10X SDS-PAGE loading buffer was added to each of the samples of protein that consisted of 100 mM Tris-base (pH=6.8), 1% SDS, 10 mM DTT, 50% glycerol, and deionized water. To determine how much sample should be loaded into each well, protein concentrations were determined spectrophotometrically using the BioRad protein identification reagent system (BioRad). The protein concentration of the samples was based on a standard curve produced. The volume of each sample amounting to 100 pg of protein was loaded into each of the wells for electrophoresis. A kaleidoscope prestained protein standard (BioRad) was loaded into one of the wells on each gel. The gels were run at 200 volts until the bands showed distinct separation. Western Blot Rabbit antibodies against phosphorylated-Akt—Z, Akt-2, IRS-1, and p7OS6K and phosphorylated-p70 S6K were obtained from Cell Signaling Technologies. Positive control cell extracts for Akt, phosphorylated-Akt, p70 S6K, and phosphorylated-p70 S6K were used and obtained from Cell Signaling Technology. These were also loaded into the wells of the gel with the samples. The controls were used not only to validate antibody detection, but were also used to normalize detection levels among the proteins. 41 In order to prepare for the Western blot, the SDS-PAGE gel was transferred electrophoretically to a nitrocellulose membrane. The transfer was run in a buffer consisting of 25 mM Tris-base, 192 mM glycine and 20% weight to volume of methanol. Following transfer, protein bands were visualized using Ponceau S stain. The gels were stained with Commassie Blue to visualize and confirm the transfer of protein. The membrane was incubated in blocking buffer that was constructed according to the manufacturers’ instructions for each individual antibody. The primary antibodies were diluted then applied to the membrane to incubate for 18 to 24 hours. Following incubation, the membranes were washed in 1X TBS/T then the secondary antibody was applied. The secondary antibody used was ImmunoPure peroxidase conjugated goat anti- rabbit IgG (Pierce) and was diluted 1:2000. After an hour of incubation with the secondary antibody, a detection system was applied (Amersham). The membrane was then exposed to hyperfilm and developed in order to be analyzed. Quantification of the Si gnal was accomplished by Quantity One software (BioRad) using densitometric scans from autoradiograms. This system detected the density of the signal over a given number of pixels. The number of pixels analyzed for each sample on each gel was identical. The overall density displayed was determined using a standard curve with a predetermined density scale. The values obtained from the standard curve for each Western blot were normalized to the control value in Microsoft Excel and the phosphorylated to total protein levels were determined. Blocking buffers and dilutions were constructed for each of the antibodies as follows. The primary antibodies for all of the antibodies used were diluted 1:500 in blocking buffer. The blocking buffer utilized for Akt-2, p70 S6 kinase and IRS-1 42 antibodies consisted of 5% BSA, 0.1% Tween, and 1X TBS, as suggested by the manufacturer. The blocking buffer used for phospho-p70 86K and phospho-Akt-2 antibodies consisted of 50mM NaF, 5% BSA, 0.1% Tween, and 1X TBS. The dilutions for primary antibodies were determined by running a sample Western blot that was divided into multiple segments and diluted with different concentrations of antibody. The dilution ratio used was necessary to optimize the protein signal without overly increasing the background on the blot. In order to use a membrane for multiple blots, the phosphorylated antibody was assessed first, followed by the antibody for total protein. In order to prevent cross reactions following Western blot analysis of the first antibody, the membrane was stripped of the first antibody. The membrane was stripped using a solution comprised of 0.2M NaOH incubated for 10 minutes, then rinsed in TBS/T. The respective blocking buffer was then applied for the antibody and Western blot procedure was followed. This procedure was followed for both p70 S6 kinase and Akt-2. PRELIMINARY DATA Western blot analysis Following Western blot analysis for protein, it was seen that Akt-2 phosphorylation levels varied between the muscle types. In the tibialis anterior and plantaris, the phosphorylated Akt-2 ratio decreased in the diabetic muscle. Even after the treatment to destroy insulin production, Akt-2 phosphorylation was still apparent in the soleus. This muscle had a variable amount of phosphorylation among the samples. This 43 data is displayed in Figure 10 with protein bands displayed that are representative of each group. IRS-1 level were also assessed in this study. Using Western blot analysis, it was determined in the tibialis anterior that IRS-1 protein expression levels were not altered between the control and diabetic mice (Figure 11). Four mice from each group were used to obtain these results. Representative bands from the Western blot are displayed in Figure 11 with the IRS-1 quantification depicted in the graph. Since the tibialis anterior is a larger muscle and more protein could be extracted, it was the only muscle used to assess p70 S6 kinase. Even with the use of 150 pg protein, phosphorylated p70 S6 kinase was un detected to a quantifiable extent in the control or diabetic muscles. Total p70 S6 kinase was readily detectable and did not vary between the two groups. Tibialis Anterior Plantaris Soleus Ctrl STZ Ctrl STZ Ctrl STZ p-Akt —-> w “mm iriii‘lm “ m w M Akt —» mm», mm M M W W Ratio of phosphorylated Akt-2 to Total Akt-2 25 Q: 20 - 32 < i '6 15- E IControl 3' 10 IDiabetic A. .2 g 5 * o .i Tibialis Anterior Plantaris Soleus Figure 10. Western Blot Analysis: Akt-2. The average ratio of phosphorylated Akt- 2 to the total Akt-2 in each muscle is expressed in the graph The Western blot displayed is representative of the average for each group. The tibialis anterior and plantaris reached statistically significant levels between the control and diabetic (*) signifies statistical significance (p50.05). 45 Control Diabetic IRS-1 Protein Levels 35 30— 25- 20' IRS-1 Expression (Scan Units) Control Diabetic Figure 11. Western Blot Analysis: IRS-1. There was no significant difference between the control and diabetic muscles when assessed in the tibialis anterior. The plantaris and soleus muscles were not assessed. The Western blot depicted above the graph is representative of the results depicted graphically. The graph consisted of four muscles per group. 46 Tibialis Anterior Ct r| STZ Ctrl STZ p70 36k ——* ,n we” 3;;- :3 Figure 12. Western Blot Analysis: p70 S6 kinase. The phosphorylated signals were too low for quantification (top). The expression of the total p70 S6 kinase is visible in all of the samples (bottom). 47 PRELIMINARY CONCLUSIONS Specific Aim 11 contains preliminary data for Akt-2, IRS-l and p70 S6 kinase. Completion of these experiments will suggest whether critical components of the insulin signaling pathway are affected by STZ diabetes at an early time period. It would also suggest that although insulin signaling may be compromised with STZ, components of the pathway may be influenced by other proteins converging on the cascade. Furthermore, completion of these experiments would suggest whether there is a difference between signaling protein activation among muscle types during STZ diabetes. When Akt-2 is activated by phosphorylation, it is able to modulate the activity of a multitude of signaling components. The muscles used in this study displayed variability in the Akt-2 phosphorylation ratio. The dramatic increase in atrogin-l mRNA expression, as shown by the tibialis anterior in Specific Aim I was complimented by the decrease in Akt-2 phosphorylation. The plantaris muscle also followed this trend. Since the soleus showed a greater biological variability in the phosphorylation level of Akt-2, the results were more difficult to decipher. This muscle did not display a significant difference in phosphorylation from the control to the diabetic samples. Likewise in Specific Aim 1, atrogin-l was not significantly induced in the soleus. Atrogin-l expression corresponded to levels of Akt-2 phosphorylation which is consistent with the literature. As a pivotal component in the insulin signaling cascade, one explanation for the decrease in Akt could be attributable to the decrease in insulin levels that occurs with STZ diabetes. Although the insulin levels were not evaluated in this study, this STZ protocol has been shown to eliminate B cell insulin production within four days of drug 48 injection (Pighin et al. 2005). Therefore if insulin signaling is depleted, the signaling cascade beginning with the insulin receptor may be downregulated in activity. The IGF-l (Insulin-like growth factor-1) receptor also converges onto the PI3K/Akt pathway. Decreases in local or systemic levels of IGF-I may influence the phosphorylation state and activity of Akt. Since the IGF-l signaling pathway is dramatically interconnected with the insulin signaling pathway, the possibility that IGF-l is contributing, at least in part, to muscle atrophy cannot be excluded. The role of insulin and IGF-1 receptor signaling is need of further study to determine the initial signal in the development of muscle atrophy in diabetes. Pathways such as the NF-KB path also converge onto Akt, however, this is not implicated in the onset of atrophy via diabetes in skeletal muscle (Kandarian et al. 2006). Considering the data presented here, Akt phosphorylation states are clearly altered in the plantaris and tibialis anterior as a result of STZ diabetes. Conversely, the Akt-2 phosphorylation ratio was unaltered in the soleus muscle. The difference in Akt-2 phosphorylation between the muscles presented here could be attributable to many sources. One possibility for the difference in Akt-2 phosphorylation between the soleus and plantaris is that it is a result of the fiber composition of the muscle. The soleus is comprised of predominantly slow twitch fibers, while the plantaris is mostly fast twitch. Since both of these muscles are located in the posterior compartment of the hindlimb, they should experience sirrrilar patterns of contractial stimulation. Thus fiber type seems to be a greater contributor to the difference in Akt-2 phosphorylation and atrogin-l expression. A major difference between glycolytic and oxidative muscles is the ability to use fatty acids as a source of energy. As an oxidative muscle, the soleus is capable of using fatty acids for energy more readily than the 49 plantaris (Fewell et al. 1995). Finally, the difference in results between muscle types could be reflective of intrinsic concentration of specific signaling proteins. The soleus muscle has a higher intrinsic expression of IRS-1, Akt, and PIBK than the extensor digitorum longus or the epitrochearis, two predominantly glycolytic muscles (Song et al. 1999 Mar). It is unknown how much a signal must decrease before influencing its downstream target. It is plausible that the soleus is able to sustain signaling longer than the tibialis anterior and plantaris muscles due to the inherent content of Akt. At a further time point in this model, the phosphorylation ratio of Akt in the soleus may also decrease with a concomitant increase in atrogin-l mRNA expression. Furthermore, different isoforms of Akt exist. The isoform used in this study was Akt-2, however, the specificity of the antibody for this isoform may have interactions with the Akt-l isoform. More research is necessary to determine the absolute difference among the Akt isoforms and their antibody specificity. IRS-1 has been identified as a critical signaling component at the top of the PI3K/Akt cascade. In the preliminary data, phosphorylation levels of Akt-2 decreased among the plantaris and tibialis anterior muscles. Since IRS—1 is a key regulator of signal transduction to Akt-2, this protein was assessed. The Western blot exhibited an abundance of nonspecific binding, preventing complete analysis of this protein. Information can be divulged by considering the total IRS-1. Exposure to free fatty acids and hyperosmotic stress, such as hyperglycemia, causes degradation of IRS—1 (Gual et al. 2005). Therefore, an apparent decrease in IRS-1 protein could lead to a decrease in phosphorylation of Akt-2. It is unknown to what degree IRS—1 levels would need to decrease to negatively influence signal transduction to Akt-2. In this preliminary data, 50 IRS-l levels were not altered between the diabetic and control tibialis anterior muscle samples. However, Akt-2 phosphorylation levels decreased in this muscle. Several explanations count account for this data. One explanation for the results obtained is that the time period examined in this study may not be long enough for degradation to occur. Like Akt-2, IRS-l is a main component of the IGF-l receptor signaling cascade. This pathway could continue to allow stimulation and prevent degradation of IRS-l. This explanation would also require that another signal is causing a decrease in phosphorylation of Akt-2. Once again, assessment of IGF—1 during STZ diabetes would provide further answers. Another interpretation of these results is that although the degradation is unaffected is this preliminary data, perhaps the protein is sterically inhibited preventing the signal to be tranduced along the cascade to Akt. O-linked glycosylation by hyperglycemia is one mechanism that causes inhibition of IRS-1 (Gual et al. 2005). In this study, the diabetic mice exhibit hyperglycemia making it conceivable that hyperglycemia is causing inhibition of IRS-1 in the diabetic mice preventing phosphorylation of Akt while maintaining protein expression. Degradation and steric hindrance may take a longer time period to influence IRS-1 than the 7 day time point. Taking these possibilities into account, perhaps Western blot analysis is not an appropriate measure of IRS-1. Measuring IRS-l activity may offer more of an explanation into the control of this molecule in STZ diabetes. Again, experimenting with amelioration of hyperglycemia would offer further assessment of the roles of insulin and glucose in this signaling cascade. 51 Another complication with this preliminary data could be attributable to the specificity of the antibody. Even with repetition of the Western blot, IRS-l exhibited an abundance of unspecific binding. It would be appropriate the use IRS-l antibodies from other companies or different lot numbers to determine the actual levels of IRS-1 expression. Another possibility is that the antibody is identifying IRS-1 degradation products. Furthermore, the Western blot displayed in Figure 11 assesses only four tissue samples. Using a greater number of samples may show that there is a difference between the two experimental groups. Further experimentation is necessary to determine the complete effect of STZ diabetes on this signaling molecule. Considering the data from the p70 S6 kinase phosphorylation levels would allow protein synthesis rates to be evaluated. Since p70 S6 kinase is partially regulated by Akt- 2 phosphorylation, it would be expected that p70 S6 kinase phosphorylation would decrease when phosphorylation ratio of Akt-2 also decreases. In this preliminary data, however, the phosphorylation levels were unable to be assessed. Since the protein concentration of the tibialis anterior extract was larger than the soleus and plantaris, a greater quantity of protein could be loaded to the gel. This problem could be solved by concentration of the protein samples. Even after increasing the amount of protein loaded onto the gel and increasing the dilution ratio of the primary antibody, the signal produced on the blot was not quantifiable (Figure 12). The binding affinity of the antibody could also account for the inability to quantify the Western blot signal. The plantaris would be expected to follow the same trend as the tibialis anterior, as it has for Akt phosphorylation and atrogin-l mRNA expression. Total p70 S6 kinase protein 52 expression was apparent and seemingly similar between the two groups. To fully assess this data, a loading control would need to be evaluated. According to the literature, p70 S6 kinase phosphorylation levels are increased following exercise regimens. The phosphorylation levels of this protein are able to be assessed in rats following exercise protocols, however, are not as easily determined in mice. Since the rrrice in this study remained at normal activity level and were not being overly stimulated by exercise, it is a possibility that p70 S6 kinase would be at a resting level. Furthermore, determination of p70 S6 kinase phosphorylation has not been fully characterized in muscle during early administration of STZ diabetes in mice. An explanation for the inability to identify a signal could be due to a basally low level of phosphorylated p70 S6 kinase in addition to our search for a diminishing signal. If p70 S6 kinase phosphorylation would have been determinable in this study, it would have lead to several possibilities. Under the assumption that p70 S6 kinase phosphorylation was able to be assessed, like Akt-2, the phosphorylation may differ among the muscles. If the phosphorylation was increased in the control over the diabetic, it would appear that p70 S6 kinase would follow the phosphorylation of Akt. One interpretation of these results would be that p70 S6 kinase is being activated by an alternate path. This would be consistent with MAPK activation of p70 S6 kinase, under the assumption that MAPK is activated differently during STZ diabetes (Markuns et a1. 1999). Another interpretation would be that although Akt is decreased in phosphorylation, it may need a further reduction in phosphorylation before impacting mTOR signaling. Since mTOR is in direct regulation of p70 S6 kinase, any effector 53 modulating mTOR could impact the phosphorylation levels of p70 S6 kinase. The phosphorylation levels of mTOR would be helpful in assisting this analysis. As determined by the preliminary data in Specific Aim 1, atrogin-l is upregulated with type I diabetes. Since it is necessary for FOXO transcription factors to bind to the atrogin-l promotor in order to increase mRNA expression, this data suggests that FOXO transcription factors have been activated with early STZ diabetes. Collectively, these results indicate the complexity of the insulin signaling pathway. Based on the preliminary data and anticipated results of continued research, it may indicate that there are many other factors contributing to overall regulation of the insulin signaling cascade. Each of these pathway constituents are influenced by other signaling molecules within the cell. In order to examine a single pathway, the components must be examined individually, however, consideration of other effectors must be acknowledged. More research is necessary to fully determine which signals are altered in STZ diabetes and are important to the onset of muscle atrophy. CONTINUED RESEARCH Insights into how muscle atrophy signals are initiated by signaling components in STZ diabetes will be obtained from the conclusions of the aforementioned experiments. In order to fully reveal the impact of STZ diabetes on skeletal muscle, further study is necessary. In this project, mRNA levels of 4E-BP1 and caspase-3 protein activation could be determined, as well as assessing IRS-1 and p70 S6 kinase to completion. Myofibrillar breakdown could be measured as activation levels of capase-3. Caspase-3 is responsible for degradation of myofibrillar protein which enables the 54 fragments to be further degraded by ubiquitin li gases. We would anticipate that Western blot analysis of caspase-3 and cleaved caspase-3 show in the tibialis anterior that cleaved caspase-3 is abundant. This would correspond to the increase in muscle atrophy that has been depicted in this study. The soleus and plantaris muscles would potentially display decreased cleavage of caspase-3 in comparison to the tibialis anterior. The plantaris, however, would show more activation than the soleus muscle. Overall, this data would indicate whether breakdown of myofibrillar protein was occurring early in STZ diabetes. In this project, mRNA levels of 4E-BP1 could also be determined. It has been shown that mRN A levels of 4E-BP1 rise with chronic atrophy and is another factor controlled by the Akt/mTOR signaling pathway (Kandarian et al. 2006). This would provide further insight into the activity of protein synthesis during loss of insulin signaling. Overall it is apparent that further efforts to understand the mechanism behind the response elicited by STZ diabetes are needed. These should entail further investigation with components that impact the insulin receptor signaling pathway. As in Specific Aim 1, earlier time periods could be examined to determine when signaling components are altered during atrophy. Some of these factors could include determining phosphorylation of the insulin receptor itself, PIBK, FOXO, or mTOR. Further investigation of the FOXO family also needs to be examined in order to determine precisely what each isoform regulates. Continued repetition of PCR and Western blots should be run as additional animal trials with STZ diabetes conclude. More samples would provide more statistically 55 relevant data. These data will increase our understanding of the mechanism diabetes might use to provoke muscle atrophy and further complications. 56 SPECIFIC AIM III INTRODUCTION Thus far, this project has shown that disruption in glucose homeostasis by STZ causes skeletal muscle atrophy to ensue. PGC-la is a transcriptional coactivator that responds to physiologic stressors, allowing the cell to meet the changing energy demands. This component involved in metabolic regulation may also play a role in muscle atrophy by interacting with components of the insulin signaling cascade. Therefore, this specific aim seeks to determine if PGC-la expression is altered with STZ diabetes. It can be hypothesized that when diabetes is induced pharmacologically by STZ, PGC-la expression levels decrease. Furthermore, PGC—la expression levels will vary between muscle types since different amounts of weight loss were visible depending on the muscle. Support for this hypothesis is gained from the report by Patti et al. (2003) that reveals decreased PGC-la expression in type H diabetes mellitus. This provides evidence for altered control of PGC-la due to metabolic imbalance in the diabetic state. In this study, the authors did not address multiple muscle types. This is of importance since overall PGC—la expression may vary between muscles. Mootha et al. (2003) also showed during type H diabetes that PGC-la regulated genes were coordinately downregulated in muscle. These reports did not investigate PGC-la using multiple muscle types nOr did they inspect PGC-la expression during a period of early atrophy. Since muscle wasting decreases force production and increases the vulnerability of muscle to injury, other proteins may act to prevent the production of muscle wasting. 57 Sandri et al. (2006) showed prevention of atrogene upregulation with overexpression of PGC-la. They determined this was attributable to PGC-la interaction with FoxO3. Their study paints a protective role for PGC-la in muscle atrophy. PGC—lu has been well investigated following exercise, but its expression during early STZ induced muscle atrophy has not been thoroughly investigated. This section seeks to explore whether there is altered control of the energetic regulator, PGC-la, during metabolic imbalance and muscle atrophy induced by STZ diabetes. MATERIALS AND METHODS Animals For all proposed experiments, the mice should be treated as they were in the materials and methods of Specific Aims I and II. Quantitative Real Time PCR For this experiment, quantitative real time PCR was carried out as described in Specific Aim 1. The primer to be used for PGC-la was obtained from Invitrogen. The forward and reverse sequences are listed in Table 1. Enzyme Assay Mitochondrial integrity was assessed using a citrate synthase enzyme assay. For this assay, the muscle portions were homogenized in a 0.05 M Tris HCl buffer (pH=8.0). The fractions were weighed and 1 part muscle for every 20 parts buffer was added to the homogenization via]. The tissue was homogenized for 30 seconds with the Ultraterrax homogenizing tool at a setting of 3. The homogenate was then centrifuged for 3 minutes 58 at a speed of 900g at room temperature, followed by removal of the supernatant. Homogenizing buffer was added to reach a final dilution of 1:400. 100 mM Tris buffer (pH=8.0), 0.1 mM DTNB, and 0.5 mM oxaloacetate were mixed and allowed to equilibriate for 5 minutes. The homogenate (20 ul) was added to the mixture as well as 0.3 mM acetyl CoA whereby the rate of mercaptide ion production was measured spectrophotometrically at 412 nm. This method of citrate synthase activity was measured according to the method of Srere (1966). The protein concentration was used to normalize the activity of the citrate synthase enzyme. PRELIMINARY DATA The basal mRNA expression levels of PGC—la in the controls did not vary between muscle types (Figure 13). The tibialis anterior and plantaris muscles did not differ in PGC-la expression between diabetic and control samples. The slow twitch soleus muscle exhibited significantly decreased levels of PGC-la in comparison to its control. The tibialis anterior showed a large degree of variability in PGC-la expression. The expression of PGC-la was expressed as relative to the expression of B—actin. Mitochondrial activity was evaluated by enzymatic analysis of citrate synthase. Results from the tibialis anterior at 5 and 17 days showed no difference inactivity levels. Due to experimental design, the soleus and plantaris were unable to be assessed. 59 PGC-1 mRNA Expression 01 h I Control I Diabetic PGC-1 expression (A U) M co _A 0 % Tibialis Anterior Plantaris Soleus Figure 13. PGC-la mRNA Expression. The diabetic tibialis anterior reached a statistically different level of PGC-lo. expression. There was no difference between the plantaris and the soleus muscles. PGC-la mRNA expression is in arbitrary units (A U), (*) indicates statistical significance. 60 PRELIMINARY CONCLUSIONS The framework for Specific Aim HI was built on the decreased expression of PGC-la evident in type II diabetes and the potential ability of this protein to act as a deterrent to muscle atrophy (Patti et al. 2003; Sandri et al. 2006). Diabetes mellitus insti gates an imbalance in metabolic control, pointing towards an influence on PGC-la regulation. The preliminary data presented in this Specific Aim show that PGC-la expression levels are altered by STZ diabetes in the soleus muscle, but not in the tibialis anterior or plantaris. The slow twitch soleus muscle was shown to have a decrease in levels of PGC-la mRNA expression, but not measurable weight loss in the muscle (Figures 11, 3). The tibialis anterior, however, did show measurable weight loss and activation of atrogin-l without a change in PGC-la (Figures 3, 8, 11). This preliminary data proved to be contradictory to our original hypothesis. There are several explanations for the results that have been obtained from this preliminary data. In previous studies using the same streptozotocin protocol, citrate synthase enzymatic activity was assessed in order to determine whether the capacity of the mitochondria was affected. This previous study revealed in the tibialis anterior that citrate synthase activity was unaltered 5 and 17 days following STZ treatment. This indicates that the density of the mitochondria was not altered in this diabetic model, validating the results establish here with PGC—la expression. Oxidative muscle, however, was not examined in the previous citrate synthase study. From the PGC—la data divulged above, it seems that the soleus would ultimately acquire a decreased ability for mitochondrial biogenesis and lessened oxidative capacity in comparison to the 61 control. The glycolytic muscles, however, do not seem to shift towards a decrease in ability for oxidative capacity at this time point. In response to exercise-induced upregulation of oxidative genes by PGC-la, muscle type specificity is seen (Ikeda et al. 2006). This is partially consistent with the preliminary data presented here in the display of varied expression of PGC-la by muscle type. Exercise notoriously increases the expression levels of PGC-la in skeletal muscle (Handschin et al. 2003). With consideration of this present study, the activity levels of the mice were not greatly altered among the control and diabetic mice. Therefore, the major difference appears to be the metabolic imbalance caused by diabetes which includes increased blood levels of fatty acids and glucose, with decreased insulin. This suggests that one or more of these factors is ultimately causing the expression of PGC-la to become altered. Metformin treatment, a drug that decreases hyperglycemia, caused an increase in PGC-la levels among diabetic models (Kumagai et al. 2006). This increases the likelihood of heightened glucose as a signal influencing PGC-la expression. The muscles presented here displayed differences in mRNA expression despite encountering identical levels of circulating blood glucose. The difference among the muscles examined here could be attributable to oxidative and glycolytic phenotypes. As an oxidative muscle, the soleus showed altered PGC-la expression, while the predominantly glycolytic muscles did not. As a potential reason for this occurrence, the soleus relies on fatty acids as a source of energy more than the glycolytic muscles. Therefore, the increase in fatty acids that is elicited by the diabetic condition (Chen g et al. 2006) influences the soleus more so than the other muscles. 62 Since the soleus requires more constant energy supply than the primarily glycolytic muscles presented here, the decrease in PGC-la expression could indicate the transition of metabolic phenotypes. Feczko et al (1994) showed a fiber type transition from type 2A to 2B fibers over the course of 42 days after induction of diabetes by STZ. This demonstrates the ability of muscle to change in response to metabolic imbalance. This study does not take into account slow oxidative muscles nor an early time point. Our preliminary data presented here shows that downregulation of a transcriptional regulator of metabolic genes is altered preferentially in oxidative muscle. This may be an early indication that the soleus is transitioning from a slow to a faster phenotype. This result corresponds to fiber type switching in humans with diabetes (Oberbach et al. 2006). This preliminary data provides just a small glimpse into alterations in metabolic control during diabetes. The data presented here introduce a variety of potential connections of PGC-la with muscle atrophy and insulin signaling components. Important to consider are the possible influences that PGC-la may have over skeletal muscle atrophy. One possibility is that interaction of PGC-la and the insulin signaling pathway involves regulation through Akt. As a coactivator of transcription, PGC-la controls transcription of genes involved in glucose metabolism and fatty acid oxidation. One of the genes controlled by PGC-lu is TRB-3, a mammalian homolog of the drosophila tribbles gene (Mortensen et al. 2006). This protein is capable of binding and suppressing Akt activity in the liver and myotubes (Du et al. 2003). Although PGC-la levels were decreased in the soleus, atrophy was not displayed in this muscle by weight or molecular markers of atrophy. Also in the soleus, Akt-2 phosphorylation was not modified, as determined in Specific 63 Aim H. In accordance with the preliminary data presented here, it is plausible that PGC- la downregulation in the soleus causes downregulation of TRB-3, which eliminates the negative regulation imposed on Akt. Meanwhile, in the tibialis anterior and plantaris, PGC-la levels allow continued expression of TRB-3, allowing Akt phosphorylation to be suppressed. This scenario would require further characterization of TRB-3 as it has not been examined using skeletal muscle from a STZ diabetic model. Further interaction of PGC-la and muscle atrophy is implicated by Sandri et al (2006). Overexpression of PGC-la prevented a rise in atrogene expression during denervation. They suggest that this occurred as a result of PGC-la interaction with FoxO3. This suggests that muscles preferentially expressing higher protein levels of PGC-la may have a greater ability to resist atrophy. Puigserver (2005) suggests that PGC-la is preferentially expressed in oxidative muscle, such as the soleus. Our data displayed a decrease in PGC-la mRNA in the soleus, without indication of atrophy. Examination of the protein content of PGC-la would be helpful in characterization of this affect on atrophy. Much further research is necessary to determine the interplay of atrophy and mitochondrial biogenesis and the mechanisms that employ the components. In conclusion, STZ diabetes causes metabolic imbalance with a differential influence according to muscle type. The reasons for this effect are still in need of characterization. The oxidative phenotype shows a resistance to muscle atrophy, which may be contributed by PGC-la. More precise studies of the involvement of PGC-la in metabolic control and muscle atrophy during STZ diabetes will be required. CONTINUED RESEARCH 64 Insights into mechanisms that allow skeletal muscle to atrophy at different rates can be partially divulged from the preliminary data above. Research that builds further on that data may ask how various transcriptional activators interact in the nucleus of the muscle cell. Mutants using gene silencing techniques with various components that affect PGC-la and atrogenes could be used. Physiologic data including that of fatty acid and insulin measurements would be helpful to further identify metabolic alterations in STZ diabetes. Further investigation into earlier and later periods within the diabetic time course may provide further indications of fiber type switching. Histological data would also provide helpful to determining whether a difference in fiber type was apparent between diabetic and control muscle types. Since histology may take a prolonged period to show a visible difference in muscle fibers, mRNA expression is helpful in assessing whether fiber type switching is occurring at this early period. If this is verified even further, it may exemplify the remarkable plasticity of muscle. Instead of measuring transcriptionally activated genes of FOXO in order to determine its activation, phosphorylation of FOXO by Western blot analysis could be analyzed. This would determine the direct activity of FOXO instead of indirect assumption. Furthermore, the protein levels of FOXO according to tissue type would be helpful to determine. This could be one of the contributors to differences in muscle atrophy between muscle types. Advances in characterization of the FOXO family as a whole to determine which activators may be in direct connection with PGC-la activation is definitely needed for research in the future. Further research could also include 65 looking into the role of TRB-3 in STZ diabetes as it has not been investigated in this model to any extent. Continued research could also include taking a look into the impact of diabetes on more muscles, including cardiac muscle. As the heart is extremely oxidative, it could provide further insight into the actions of PGC—la in muscle. Beyond atrophy initiation, the relationship between diabetes and skeletal muscle atrophy mechanisms may be uncovered through understanding of how the cell guards against muscle atrophy to begin with. This understanding may further enhance our understanding of the mechanisms of atrophy. More data and research is needed to determine what controls PGC-la expression during diabetes. Hypoinsulinemia, hyperglycemia, or the increase in fatty acids within the bloodstream could potentially be the culprit of deactivation. It would be helpful to examine each to these effectors on muscle during diabetes individually. 66 DISCUSSION This work aims to clarify the relationship between STZ diabetes and muscle atrophy. The areas examined are: 1) occurrence of skeletal muscle atrophy in a model of type I diabetes 2) determinants of the insulin signaling pathway that may lead to muscle wasting and 3) impact of STZ diabetes on PGC-la expression according to muscle type during early STZ diabetes. As previously stated, the answers to these questions will shed light on skeletal muscle atrophy induced by diabetes as well as offer clues for a mechanism of atrophy in other sources. Original preliminary data along with data reported so far by other research groups support our hypothesis that metabolic disruption by STZ diabetes plays a critical role in the onset of muscle atrophy. Continued research in this area is important because it will shed light on the complications of diabetes in muscle degeneration that may be applicable to atrophy induced by other means. In specific aim I, atrophy was observed in experimental diabetes mouse models. Diabetes was induced with a single pharmacologic agent, streptozotocin. The mice that received the drug treatment had a decrease in body weight and heighten blood glucose levels. Three different muscle groups were examined for the affects of muscle wasting. Skeletal muscle atrophy was assessed by increased mRNA expression of genes known to be activated in all forms of atrophy. Figure 3 shows that the muscle weight loss occurred differentially among the muscle types. The tibialis anterior showed decrease in weight as well as activation of atrogin-l. The plantaris muscle trended toward a decrease in weight with an corresponding trend toward increased atrogin-l gene expression. The soleus did not indicate a decrease in weight or an increase in atrogin-l mRNA. MuRFl expression 67 was unaltered in all of the muscle types suggesting that it may take a longer time period to be activated or that it may be under the control of other factors. Thus, the atrophy process had been initiated in the diabetic animals, which was reflected in the decrease in total body weight. Since hyperglycemia and hypoinsulemia were the predominant alterations from homeostasis in these early animal models of type I diabetes and even when hyperglycemia is ameliorated during streptozotocin induced diabetes the animals still experience a drastic decrease in weight loss, hypoinsulinemia would seem to be the culprit causing the increase in muscle degeneration. Specific components of the insulin signaling cascade were examined in the next section. Since a vast amount of proteins have been implicated in the insulin cascade, it is imperative that research use simplified models. By using the STZ diabetic model and isolating the components associated with insulin and atrophy, a simplified model was utilized in this project. Preliminary Western blot analysis show altered phosphorylation levels of Akt with STZ diabetes. Interestingly there are also differences in phosphorylation levels between different muscle types. IRS-1 levels were unaltered in the tibialis anterior suggesting inhibition of this molecule or alterate control of Akt phosphorylation. One area of continued research here is to discern the differences between the muscles that allow for atrophy to occur at different rates. This may prove difficult since there are a multitude of differences in protein isoforms and concentrations among the muscles. The main focus of this continued research should be to gain more detail about the insulin response through quantitative real time PCR and Western blot analysis. 68 A main difference between the muscle types is their ability to rely on glycolytic or oxidative energy production. In order to investigate one protein that is be responsible for controlling cellular metabolism and may have a role in atrophy, PGC- l or expression was determined. Other components may converge on the insulin signaling pathway to determine overall atrophy in muscle. PGC-la expression has been shown to decrease with type H diabetes, suggesting altered regulation of this protein (Patti et al. 2003). It has been proposed by Speigelman et al (2006) that PGC-la inhibits FoxO3 upregulation of atrogin-l expression during contractial stimulation. Similar experimentation using type I diabetes as a model when insulin is obliterated has not been examined. As well as FOXO interaction, PGC-la is a major contributor to the expression of genes involved in oxidative metabolism. It causes upregulation of TRB-3, a protein that is able to suppress Akt activity. These studies offer further support for a role of PGC-lor in skeletal muscle atrophy via the insulin signaling components. As in type II diabetes, PGC-l levels were altered with STZ diabetes. In Specific Aim III, PGC-la levels decreased only in the soleus following induction of diabetes. The preliminary data looking at PGC-la may indicate a trend towards fiber type switching at an early period of diabetes. Feczko et al. (1994) have shown with STZ diabetes that fiber type switching does occur over an extensive period of time. These preliminary results may indicate that gene expression is apparent before visible and functional changes in fiber types can be determined. Repeated animal models should be set up and PCR run at earlier time points in the atrophy process to determine whether fiber type switching is in the beginning stages at this early time point. 69 The preliminary data indicates the intricacy of muscle atrophy in early STZ diabetes. Many other factors still need to be examined. This study indicated the importance of the muscle model used when experimenting with muscle atrophy as each muscle has distinct characteristics and reactions that need to be considered. CONCLUSIONS Insulin signaling is essential to normal homeostasis of skeletal muscle and is disrupted during diabetes. Insulin regulates not only uptake of glucose as energy for the muscles, but impacts protein synthesis and degradation pathways in the muscle. Overall, the preliminary data collected here support the hypothesis that STZ diabetes upregulates atrophy processes in skeletal muscle. Our preliminary data also shows that the different muscles experience different rates with this diabetic model. Components of the insulin signaling pathway are implicated in causing protein degradation and synthesis. In this project, phosphorylated Akt, a major signaling protein, was altered with STZ diabetes. The literature establishes that PGC-la may play a preventative role in muscle atrophy and also that PGC-la expression levels decrease during type II diabetes (Sandri et al. 2006; Patti et al. 2003). The preliminary data show that STZ diabetes influences components of the insulin signaling pathway and expression of P601, but further research is needed to determine the regulatory mechanism. Oxidative and glycolytic muscles respond differentially to the induction of diabetes by STZ. PGC—la levels may control the expression of genes necessary for metabolism and consequently cause skeletal muscle to atrophy at difference rates due to interaction with the insulin signaling cascade. Further research is necessary to elucidate the complete mechanism of muscle atrophy during STZ diabetes. 70 CLINICAL SIGNIFICANCE The results of this work hold clinical value in determining how the loss of insulin signaling affects skeletal muscle. Since atrophy is also implicated in many other forms of disease as well as inactivity, this research holds answers to many unanswered questions in atrophy. Many forms of atrophy are characterized by similar biochemical changes (Lecker et al. 2004). Therefore using this model, advances applicable to muscle wasting induced by other means can also be elucidated. Further clinical benefits will be obtained if the mechanism of atrophy due to loss of insulin signaling such as in type I diabetes becomes clearer. Prevention and treatments will follow providing an understanding of the disease mechanism. 71 BIBLIOGRAPHY Antonetti DA, Reynet C, and Kahn CR. (1995) Increased expression of mitochondrial- encoded genes in skeletal muscle of human with diabetes mellitus. Journal of Clinical Investigation. 95:1383-1388. Armstrong RB and Ianuzzo CD. (1977) Compensatory hypertrophy of skeletal muscle fibers in streptozotocin-diabetic rats. Cell Tissue Res. Jul;181(2):255-66. Aughsteen AA, Billah Khair A, and Suleiman AA. (2006) Quantitative morphometric study of the skeletal muscles of normal and streptozotocin—diabetic rats. J Pancreas 7(4):382-389. Baldwin KH, Herrick RE, Ilyina-Kakueva E and Oganov VS. (1990) Effects of zero gravity on myofibril content and isomyosin distribution in rodent skeletal muscle. FASEB J 4:79-83. Beck- Nielson H, Vaag A, and Damsbo P. (1992) Insulin resistance in skeletal muscle in patients with NIDDM. Diabetes Care 15:418-429. Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA, Poueymirou WT, Panaro FJ, Na E, Dharrnarajan K, Pan ZQ, Valenzuela DM, DeChiara TM, Stitt TN, Yancopoulos GD, and Glass DJ. (2001) Identification of ubiquitin ligases required for skeletal muscle atrophy. Science. Nov;294(5547):1704-8. Bouillon R, Bex M, Van Herck E, Laureys J, Dooms L, Lesaffre E, and Ravussin E. (1995) Influence of age, sex, and insulin in osteoblast function: osteoblast dysfunction in diabetes mellitus. J Clin Endocrinol Metab. 80:1194-1202. Bouzakri K, Zachrisson A, Al-Khalili L, Zhang BB, Koistinen HA, Krook A, and Zierath JR. (2006) siRNA-based gene silencing reveals specialized roles of [RS-1/Akt2 and IRS- 2/Akt1 in glucose and lipid metabolism in human skeletal muscle. Cell Metab. Jul;4(1):89-96. Cao PR, Kim HJ, and Lecker SH. (2005) Ubiquitin-protein ligases in muscle wasting. Int. J. Biochem. Cell Biol. 37:2088—2097. Cardozo T and Pagano M. (2004) The SCF ubiquitin ligase: Insights into a molecular machine. Nature Reviews Molecular Cell Biology. 5:739—751. Charron MJ, Brosius FC, Alper SL, and Lodish HF. (1989) A glucose transport protein expressed predominately in insulin-responsive tissues. Proc Natl Acad Sci U S A. Apr;86(8):2535-9. 72 Chen V and Ianuzzo CD. (1982) Dosage effect of streptozotocin on rat tissue enzyme activities and glycogen concentration. Can J Physiol Pharmacol. Oct;60(10): 1251-6. Center for Disease Control, (2005) http://www.cdc.gov/diabetes/pubs/estimates.htm#prev Centner T, Yano J, Kimura E, McElhinny AS, Pelin K, Witt CC et a]. (2001) Identification of muscle specific ring finger proteins as potential regulators of the kinase domain. J Mol Bio 306:717-726. Cheng JT, Huang CC, Liu 1M, Tzeng TF, and Chang CJ. (2006) Novel Mechanism for Plasma Glucose—Lowering Action of Metformin in Streptozotocin-Induced Diabetic Rats Diabetes. Mar;55(3):819-25. Chibalin AV, Yu M, Ryder JW, Song XM, Galuska D, Krook A, Wallberg-Henriksson H, and Zierath JR. (2000) Exercise-induced changes in expression and activity of proteins involved in insulin signal transduction in skeletal muscle: differential effects on insulin- receptor substrates 1 and 2. Proc Natl Acad Sci U S A. Jan;97(1):38-43. Criswell DS, Booth FW, DeMayo F, Schwartz RJ, Gordon SE, and Fioretto ML. (1998) Overexpression of IGF-I in skeletal muscle of transgenic mice does not prevent unloading-induced atrophy. Am J Physio]. 275(3 Pt l):E373-9. Crozier SJ, Anthony JC, Schworer CM, Reiter AK, Anthony TG, Kimball SR, and Jefferson LS. (2003) Tissue-specific regulation of protein synthesis by insulin and free fatty acids. Am J Physiol Endocrinol Metab. Oct;285(4):E754-62. Du J, Wang X, Miereles C, Bailey JL, Debi gare R, Zheng B, Price SR, Mitch WE. (2004) Activation of caspase-3 is an initial step triggering accelerated muscle proteolysis in catabolic conditions. J Clin Invest. J an;l 13(1):l 15-23. Du K, Herzip S, Kulkami RN, Montminy M. (2003) TRB3:a tribbles homolog that inhibits Akt/PKB activation by insulin in liver. Science 300: 1574-1577. Fewell JG and Moerland TS. (1995) Responses of mouse fast and slow skeletal muscle to streptozotocin diabetes: myosin isoenzymes and phosphorous metabolites. Mo] Cell Biochem. Jul;148(2):147-54. Feczko JD and Kleuber. (1994) Anatomical Record 239:18-34. Finck BN and Kelly DP. (2006) PGC-l coactivators: inducible regulators of energy metabolism in health and disease. J Clin Invest. 116(3):615-22. Fingar DC and Blenis J. (2004) Target of raparnycin (TOR): an integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression. Oncogene. 23:3151—3 171. 73 Gual P, Le Marchand-Brustel Y, and Tanti JF. (2005) Positive and negative regulation of insulin signaling through IRS-l phosphorylation. Biochimie 87: 99—109. Gomes MI), Lecker SH, J agoe RT, Navon A, and Goldberg AL. (2001) Atrogin-l, a muscle specific F—box protein highly expressed during muscle atrophy. PNAS. 98 (25): 14440-14445. Handschin C, Rhee J, Lin J, Tarr PT, and Spiegelman BM. (2003). An‘autoregulatory loop controls peroxisome proliferator-activated receptor 7 coactivator la expression in muscle. PNAS 100(12):7111-7116. Henriksen EJ, Bourey RE, Rodnick KJ, Koranyi L, Perrnutt MA, and Holloszy JO. (1990) Glucose transporter protein content and glucose transport capacity in rat skeletal muscles. Am J Physiol. Oct;259(4 Pt l):E593-8. Hershko A and Ciechanover A. (1998) The ubiquitin system. Annu Rev Biochem. 67:425-79. Ikeda S, Kawamoto H, Kasaoka K, Hitomi Y, Kizaki T, Sankai Y, Ohno H, Haga S, and Takemasa T. (2006). Muscle type-specific response of PGC-la and oxidative enzymes during voluntary whee] running in mouse skeletal muscle. Acta Physiol. 188:217-223. J ackman RW and Kandarian SC. (2004) The molecular basis of skeletal muscle atrophy. Am J Physiol Cell Physiol. Oct;287(4):C834-43. James DE, Zorzano A, Boni-Schnetzler M, Nemenoff RA, Powers A, Pilch PF, and Ruderrnan NB. (1986) Intrinsic differences of insulin receptor kinase activity in red and white muscle. J Biol Chem. Nov;26l(32):l4939-44. Kahn R. (1994) Insulin action, diabetogenes, and the cause of type II diabetes. Diabetes. 43(8): 1066-84. Kainulainen H, Schurmann A, Vilja P, and Joost HG. (1993) In-vivo glucose uptake and glucose transporter proteins GLUTl and GLUT3 in brain tissue from streptozotocin- diabetic rats. Acta Physiol Scand. Oct;l49(2):221-5. Kamei Y, Miura S, Suzuki M, Kai Y, Mizukami J, Tani guchi T, et a]. (2004) Skeletal muscle FOXOl (FKHR) transgenic mice have less skeletal muscle mass, down-regulated Type I (slow twitch/red muscle) fiber genes, and impaired glycemic control. J Biol Chem 2004;279:41114—41123. Kamura T, Koepp DM, Conrad MN, Skowrya D, Moreland RJ, Illiopuolos O et a]. (1999) bel, a component of the VHL tumor suppressor complex and SCF ubiquitin ligase. Science 284:657-661. 74 Kandarian SC and Jackman RW. (2006) Intracellular signaling during skeletal muscle atrophy. Muscle & Nerve. Feb;33: 155-165. Kedar V, McDonough H, Arya K, Li HI-I, Rockman HA, and Patterson C. (2004) Muscle specific RING finger I is a bona fide ubiquitin ligase that degrades cardiac troponin 1. Proc Nat] Acad Sci USA 52: 18135-18140. Kido Y, Nakae J, and Accili D. (2001) The insulin receptor and its cellular targets. Journal of Clinical Endocrinology. 86(3):972-979. Lecker SH, J agoe RT, Gilbert A, Gomes M, Baracos V, Bailey J, Price SR, Mitch WE, and Goldberg AL. (2004) Multiple types of skeletal muscle atrophy involve a common program of changes in gene expression. FASEB J. Jan;18(1):39-51. Lee SW, Dai G, Hu Z, Wang X, and Mitch WE. (2004) Regulation of muscle protein degradation: coordinated control of apoptotic and ubiquitin-proteosome systems by phosphatidylinositol 3 kinase. J Am Soc Nephrol. Jun;15(6):1537-45. Like AA and Rossini AA. (1976) Streptozotocin-induced pancreatic insulitis: new model of diabetes mellitus. Science. 193:415-417. Lin J, Wu H, Tarr PT, Zhang CY, Wu Z, Boss 0, Michael LF, Puigserver P, Isotani E, Olson EN, Lowell BB, Bassel-Duby R, Spiegelman BM. (2002) Nature. Transcriptional activator PGC-la drives formation of slow twitch fibers. 418(6899):797-801. Livak KJ and Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001 Dec;25(4):402-8. Marette A, Richardson JM, Ramlal T, Balon TW, Vranic M, Pessin JE, and Klip A. (1992) Abundance, localization, and insulin-induced translocation of glucose transporters in red and white muscle. Am J Physiol. Aug;263(2 Pt 1):C443-52. Markuns JF, Napoli R, Hirshman MF, Davalli AM, Cheatham B, and Goodyear LJ. (1999). Effects of streptozotocin-induced diabetes and islet cell transplantation on insulin signaling in rat skeletal muscle. Endocrinology. 140(1): 106-1 1 1. Mas A, Montane J, Anguela XM, Munoz S, Douar AM, Riu E, Otaegui P, and Bosch F. (2006) Reversal of type I diabetes by engineering a glucose sensor in skeletal muscle. Diabetes. Jun;55:1546-1553. McCabe et a]. (2007) Unpublished observations via personal communications. McElhinny AS, Kakinuma K, Sorimachi H, Labeit S, and Gregorio CC. (2002) Muscle- specific RING finger-1 interacts with titin to regulate sarcomeric M-line and thick filament structure and may have nuclear functions via its interaction with glucocorticoid modulatory element binding protein-1.'Joumal of Cell Biology. 157: 125—136. 75 Moretti, A., Weig, H. J ., Ott, T., Seyfarth, M., Holthoff, H. P., Grewe, D., et a]. (2002). Essential myosin light chain as a target for caspase-3 in failing myocardium. PNAS. 99:11860—11865. Morino K, Peterson KF, and Shulman GI. (2006) Molecular mechanisms of insulin resistance in humans and their potential links with mitochondrial dysfunction. Diabetes. Dec;55:S9-S15. Mortensen OH, Frandsen L, Schjerling P, Nishimura E, and Grunnet N. (2006) PGC-la and PGC-IB have both similar and distinct effects on myofiber switching towards an oxidative phenotype. Am J Physio Endocrinol Metab 291:E807-E816. Nader GA. (2005) Molecular determinants of skeletal muscle mass: getting the “AKT” together. Journal of Biochemistry and Cell Biology. Oct;37(10):1985-96. National Institutes of Health: N ationa] Institutes of Diabetes and Digestive and Kidney Diseases. (2006) www.diabetes.niddk.nih.gov/dm/pubs/statistics Nemeth P and Pette D. (1993) Succinate dehydrogenase activity in fibers classified by myosin ATPase in three hindlimb muscles of rat. J Physio (Lond). 320273-80. Niederle B and Mayr R. (1978) Course of denervation atrophy in type I and type H fibres of rat extensor digitorum longus muscle. Anat Embryol (Berl). May; 153(1):9-21. Nolte LA, Galuska D, Martin IK, Zierath JR, and Wallberg-Henriksson H. (1994) Elevated free fatty acid levels inhibit glucose phosphorylation in slow-twitch rat skeletal muscle. Acta Physiol Scand. May; 15 l (1)25 1-9. Oberbach A, Bossenz Y, Lehmann S, Niebauer J, Adams V, Paschke R, Schon MR, Bluher M, and Punkt K. (2006) Altered fiber distribution and fiber-specific glycolytic and oxidative enzyme activity in skeletal muscle of patients with type 2 diabetes. Diabetes Care. Apr;29(4):895-900. Oku A, N awano M, Ueta K, Fujita T, Umebayashi I, Arakawa K, Kano-Ishihara T, Saito A, Anai M, Funaki M, Kikuchi M, Oka Y, and Asano T. (2001) Inhibitory effect of hyperglycemia on insulin-induced Akt/protein kinase B activation in skeletal muscle. Am J Physiol Endocrinol Metab. May;280(5):E816-24. Passmore LA and Barford D. (2004) Getting into position: the catalytic mechanisms of protein ubiquitylation. Biochem. J. 379:513-525. Patti ME, Butte AJ, Crunkhom S, Cusi K, Berria R, Kashyap S, Miyazaki Y, Kohane I, Costello M, Saccone R, Landaker EJ, Goldfine AB, Mun E, DeFronzo R, Finlayson J, Kahn CR, Mandarino LJ. (2003) Coordinated reduction of genes of oxidative metabolism 76 in humans with insulin resistance and diabetes: Potential role of PGCl and NRF 1. Proc Natl Acad Sci U S A. Jul;100(14):8466—7l. Pi ghin D, Karabatas L, Pastorale C, Dascal E, Carbone C, Chicco A, Lombardo YB and Basabe JC. (2005) Role of lipids in the early developmental stages of experimental immune diabetes induced by multiple low-dose streptozotocin. J Appl Physiol. Mar;98(3):1064-9. Puigserver P. (2005) Tissue-specific regulation of metabolic pathways through the transcriptional coactivator PGCl-alpha. Int J Obes (Lond). Mar;29(Suppl l):SS-9. Puigserver P and Spiegelman BM. (2003) Peroxisome proliferator-activated receptor- gamma coactivator 1 alpha (PGC-l ALPHA alpha): transcriptional coactivator and metabolic regulator. Endocr Rev. 24:78—90. Puigserver P, Wu Z, Park CW, Graves R, Wright M, and Spiegelman BM. (1998) A cold inducible coactivator of nuclear receptors linked to adaptive therrnogenesis. Cell. Mar;92(6):829-39. Phillips PJ, and Jeffries B. (2006) Gestational diabetes--worth finding and actively treating. Aust Fam Physician. Sep;35(9):701-3. Saltin B and Gollnick PD. (1983) Skeletal muscle adaptability: significance for metabolism and performance. Handbook of Physiology. Peachy LD, Ed. Oxford, Oxford University, p 555-631. Sandri, M., Sandri, C., Gilbert, A., Skurk, C., Calabria, E., Picard, A., et al. (2004). Foxo transcription factors induce the atrophy related ubiquitin ligase atrogin-l and cause skeletal muscle atrophy. Cell. 117:399—412. Sandri M, Lin J, Handschin C, Yang W, Arany ZP, Lecker SH, Goldberg AL, and Schwerzmann K, Hoppeler H, Kayar SR, and Weibe] ER. (1989) Oxidative capacity of muscle and mitochondria: correlation of physiological, biochemical, and morphometric characteristics. Proc Natl Acad Sci U S A. Mar;86(5):1583-7. Spiegelman BM. (2006) PGC-l ALPHA protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription. PNAS. Oct; 103(44): 16260-16265. Song XM, Kawano Y, Krook A, Ryder JW, Chibalin AV, and Zierath JR. ( 1999) Muscle fiber type specificity in insulin signal transduction. Am Joum Physiology. R1690-R1695. Srere PA. (1966) Citrate-condensing enzyme oxaloacetate binary complex. Journal of Biological Chemistry. 241:2157-2165. 77 Stitt TN, Drujan D, Clarke BA, Panaro F, Timofeyva Y, Kline WO, Gonzalez M, Yancopoulos GD, Glass DJ. (2004) The IGF-l/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin li gases by inhibiting FOXO transcription factors. Mol Cell. 14(3):395-403 Suwa M, Egashira T, N akano H, Sasaki H, Kumagai S. (2006) Metformin increases the PGC-lalpha protein and oxidative enzyme activities possibly via AMPK phosphorylation in skeletal muscle in vivo. J Appl Physiol. 101(6):]685-92. Szkudelski T. (2001) The mechanism of alloxan and streptozotocin action in B cells of the rat pancreas. Physio Res. 50:537-546. Thomason DB, Herrick RE, Surdyka D, and Baldwin KM. (1987) Time course of soleus muscle myosin expression during hindlimb suspension and recovery. J Appl Physiol. Jul;63(1):l30-7. Tintignac LA, Lagirand J, Batonnet S, Sirri V, Leibovitch MP, and Leibovitch SA. (2005) Degradation of MyoD mediated by the SCF (MAFbx) ubiquitin ligase. J Bio Chem. 280(4):2847-2856. Virkamaki A, Ueki K, and Kahn CR. (1999) Protein-protein interaction in insulin signaling and the molecular mechanisms of insulin resistance. J Clin Invest. Apr; 103(7):93 1-43. Weinert S, Bergmann N, Luo X, Erdmann B, and Gotthardt M. (2006) M line-deficient titin causes cardiac lethality through impaired maturation of the sarcomere. J Cell Biol. May;173(4):559-70. Zierath JR, Krook A, and Wallberg-Henriksson H. (2000) Insulin action and insulin resistance in human skeletal muscle. Diabetologia. Jul;43(7):821-35. Zong H, Ren JM, Young LH, Pypaert M, Mu J, Bimbaum MJ, and Shulman GI. 2002 (Dec 10) PNAS Vol 99 no 25. AMP kinase is required for rrritochondria] biogenesis in skeletal muscle in response to chronic energy deprivation. 78 uiijijrjjijjjriimini ..A. ‘1“‘