VVrrJBViJVVVV: I i 33... 30‘... 3! t I :4: V L3.oo.b§n.29iu:l .I... 1 1" ...... V. 3-. ...-9.... - tool! Vuhmflflmnfiwiraf “Kata“... J. ... I. . V V .38... V». 8. . . V VVVIQ?2V-...!’..:C~.:t&.. :0 ......‘V . V 30.0.10..- .V. V V... .9L.u0¢!( V V V ...-...}: V V V. V V V. V ..V V . V . V . V V V-.\¢.V: Vi. 0. V . ...:3h ...-.75.. . V V ..VVtVo.V.I:. iv... 3.... ...: . V V ....oV¢VJ~.'V.". 350‘1.o'-.Q¢¢>.h.' .{n v... la.-."3..s.}oo. ‘0. . ... ’353..$.V-.!.I’1: o t...- 848.133.," wgcd::¢.o.o"‘ . V «1-40-3430... V V . on... '3' ‘xu... . . . . . ... loo. 3:! 1 V . V ..3~.’DV.V' ...u. )3..;V..‘;o:’o ...". V5...- V sooa"'J-o53'v€‘ . (no. ‘.oi..oo.- zilzzl... 1.2.51.1! . V V a. V I! ‘ “O V V ' u . n V ..ths.‘ 00., V o n . . Coo-a3 Ono—...“. .JJV‘ I. V V V fiuVol€Oan¢IIVVoVY (-02: V V . V V V V V V V V .39. .- V99...V1v~ .....oV!.VIV 713 V d. ‘1..va . V . 30:0... I; V .- I ......(f V. V V V. V V V V V ‘59.... 0... anti... 0 .I-V. . 72......) oV‘QVVSI. V V . V. . V V V V 9:10.33 . Vii-...? 0” 2r!.- 1.00.83... ... V .V 1.7%. P '63. V V?! V .‘U..“V‘ Q. 0 oil. naI‘I... I... :IVOVQ“, ... v. ....‘f‘oo ... 0.730.... 3.133;: .... 733......1.‘ 0‘: 30.23733. ...:83... 1 ‘09-‘00; .. .13.... '9- .uvosV 0. V .....Voc (9!. .41313‘. .‘rV. . ... rutqu-‘Iv... it: 3.4. Vi. (‘3..31: 32c. .V... 53:!“ . ...‘lV.L-ta.o...o: 2). 0. V V . V . V. V V V. .V V V V . o‘OIOV‘o-i. V . V V N . . V V . ..nV. V V. . V V V . V 9.0. V‘ V .V V V. V V . . V 9 .2' V V lot-.5302! .53113... ...... V 9.. v .\V V V. 111.03. I..V.l.-:‘..VO‘. . V. . ...?!- 3 .0. ‘7 G... 32:11.33: . . y. 71¢..n. . V . VV V V . V I. VVSVVVI...I: .....u .5 . V . V V V . V . V . . V V V. V V . . 'Q . V ... 0.: ...-... ..- VcV ‘0. IoV- V V . .4: V V V V V V . V V V V V V V. V V 9.. y ..OVVOVH’. "37‘. V V .0". u ‘0 VI- . . V . V. V . . . . . V V V V V . ...I’. . 0‘, ...... 9". Vt:"vtkr: . ......V . .V . . . . _ . . _ . . . .V V. V V. . . . .1.» ..VV. ...VVVVV .VVVVVtVVV:VVVn.VVb.V «.... V V ... ......t V V. V V. . . V V V V :1 s. V . ach‘.\ .I Q oltLMAVVonozunruuahrt 9.5.0 V7.00..V.Q.I¢. . V V . V . . V. 2...)?! .5... HQ»... :33...‘ 3:0? .....oJV .V ....o V V . V n .....I'...V|..JIV V . V . V V V V V .V . . V. .V..¢V74.V&. w 3 ”a .3. V V V V V . V V V V V V V . . . V V V V ....V. V0...V’!oV-.9 V n .0 0‘; V. V V . V V V. VV V V V V V V V V. V .... V . ....4. V . 5.3.1.0643 V V.. .V V V . V . . V. V V _ V . V 7... ..." ...-J'u‘VO.‘ 3" .V... :1 .. ...... IVOVova’ ;_ . V V no.0...V1o‘iV ‘Vta 6:. . V ‘V... . V o. o. . V. . V V V V VV ‘.V:0Vo..V9‘V: . V V V V _ V V . V V V ’2' V‘In" V . V . V V V . V V V ...-(V't‘V 1.93... V V. V. V V V V V . . .V V V .VV .‘ fu‘oc. V . .. V . V V . V (...-.03... V V. V 0 ‘V . Q . .. ...-... o . I .V ....o a ....u.‘... o..‘.... .0 V . the... 5......VOA '00 .0 0o! .00 .85.. V7.14 a... . 3 .1...c._‘V>V.VoVV‘OSV r.-Vl..»...o p . 3.. V . V 3...... 24.90....VV V V V..- .q. v :0 0.. u . ,Q,’ E .....L: 0V0 V V V .V .V . . 5.. V . V V V ... V . «V n . V . V V V .0. I .10... V V V V V V V V . . r. 0.. . V . . V... V V V .‘-....’0 . . V ..ll-V—On7. . . vvh V V o“- . 5. V '03:»... ..T‘. .....V V...VOV"o-X:..:f7 >V VV..VVV V . ... .- 1.1.3.31. V... V. 9...... . ... ..toon. ...... ...... ..‘n.,s 0.9...- V I. :1... .th‘... ...: V. 0...... . ...-91...... 13...... o. .0 . 10...; £vu. 99.0; O...n.n.fi.”‘.9\la‘. I“ .Von"... V . u :0. . V V L c v V a..." V . V V V Vv‘ V V. V.. .‘.o V V V V V V.. V . ... . . . V V ...... .31.... ...... V - V V. .v . V V V V V . V V ......V . . V . a . V . ~ 0. V V ' V I . V ~ . c . . V V 1. o A . :45-.. V a V V ..J ..Io\- 0...: o a V . . 0‘ . V..' ...-7‘.- ..A V .V. c... ’78. O. V... . . V .V V 0.. V V . VVVQ’QV. ....VV. V VV ... VV . -.OV¢I9 V V .v0 01.. V I V. ..n. .q. .‘V 3‘. V . V V V ...... V V .V ...-2.9:). V . 5.. I. ......V v . 0 W. w 2.. n V .V V a. V on 19. - VVVIVV .V V ..o....... _ ...nl V . V . .p . .1 ¢. . .VV . D. V V V V uVQVQIIQOVo a... V. V V t. V. 0.. -.0.. o ...-o.- V V .V V . . . V V V V . V . .uut‘coit V . V . VV . . V V. V .o . V . V .V .V V V V . V. V. V .V V .VVV .V V V V VV V V V ..I. V V .. V . V. V .V V V . V V V. «V. v'cut. ...‘V V V V VVVV V V. . V V..V..-oo ..q¢f- .VVVVVV...V0.V V ...-.05.}... ...I.¢VVI 0......1. I .‘o-. .....Vo. -. V t V V .....u. lVth . a. l u A: c . . r.... .V ... V V a... g V l¢.\.vl.!. .v IOV‘IVvfi. vvvvv . xf. 10.45.. S?..~.\DVV D. . u... 'WRWV“ ...... ....VvV .....VT-oVu: to. 00.4- ....._.‘.‘o n‘...¢...vVa .ndV.V.|V4 I V V .55... u..... o . . . . .V 4 v.1. . . V V .3 V V. V V. V. V V VV . V V V V . V. V ... V V V . V ” V V. V . . .V. V. .a .‘.o.. . . v.... ...}IV V. ....l.....¢.. .. V V V V... . .V V . . VV V . .o I.- . . V 1391‘ ...~ c: 0.. V. ... ..9...I.O¢ . V V V V V V V V V V... . V V... V‘: . VV. V V. V V . . V. . V. . V. V V V . . V V V V .V V V . VQOVV . .. . V .V . . V V V V. V .. . . V .V . . V V............. V ..VV... V - ¢ 0V..s¢.oVoV . . ‘- ..V..o.9~. ...-...... . u: 1 II... .V-V . ”-9-".an O ....V of. . ‘V 6.. it . ... o J ... I‘.\ .V . V .- V. .V V V V V V V V . V . . V. V V. . V V V. . V . . V V. V. .. V. V. .V . V .....VuVaVo. V V. V .V V. . ......‘1s; .. 05.0.. h V V. z u .VV V . V ...-... V 0.9Vr' - 50" . W V . A. a V .V V V y In»... .VuV V . . V V V 7v...|l-o0n.. V VV. V V V Va... -. .0 on. V V V V V . V V V V V V V V V . V V . V V . V V V V . . V ~ 7.0.4:}... . . V V ..V w 0.0 . V . vo.V.o- .ch ...... . V n ram .. V. V. soft-V... . . til-o .. a 1m .0 I . .49. V. . . . VVY. V V V V V . V V V ... . V V V V . V.VV . V. . V V V . . V . V . . V . V V V V V _ . T316; . V 2 V .V V . V . o..V..o. ... ... V—VV" IVQQVOI‘QI.I~0 otb.t......v.%i.914.ov3vV ....(VV..X. V .V V V .V . V V V V ... V V. . V . V V V . V V V V .a-V . V V V V . VV vVVVIV 0.. V ... uVV :.vv-'§.'. 3.0.0.!an ...-lo: 0 V .‘o. 1 I V. . ... o...»o.o.v .V. . V V V V VV .V V V V V V V ... .V . V V... I... V .. V V... V. ...V . . . . V ... . ... V V V V V V. V .V .V V V V V V..s .0... .....V. n ...-i V.....V. V ... ... . V........... ... ... .su. ..Aq...s... V. O.... V.” t.....—.. o '“nvmmum .c. V ... o. . 0.9. ...‘V ..t.oo’I .0'... . ... ... V: V. V V. .....A.VuV .....‘tO... V V V V V c . .... ... V «Mm , E 0.. V V V V V V. V . . V . V V .V . . V. . V. V V V . V. V V . V VVvo . . V V . V VV V . V .....o. .. V. .I I V . V. .- - ... PM! V 1‘. . . V . ..IV‘OV .... V V V V n. . . a. ... n o . V. V V .l .. VJ... ...? 2.. ...-out... V n. 23. V .V V V. .... . V . . . V _ . V V. V . V. V V. V .V V V V V .20.... . .V V. . V V VV V .V VV . . ...-'V . V . V ...-oQVOOV- . V . V . . I. .Vol II V- ooV~Ilnonuuq V y . ...-"-‘O V ..V ....V.. V. V V V... ‘ f“ v 0 o. uj'fi! V. ....V n. .V V . AV . V V V V V .... V V V. . VV . V V. V . V . V . V V . . V .V V. .V V . V .01 V . V . V . V V0.1...‘GufionViVO V V... This is to certify that the dissertation entitled ROLE OF THE SPHINGOMYELIN/CERAMIDE PATHWAY IN DIABETIC RETINOPATHY presented by MADALINA OPREANU has been accepted towards fulfillment g of the requirements for the j Doctoral degree in Microbiology and Molecular Genetics Major Professor’s Signature // / Q17 /2+ 2 C7 /0 Date MSU is an Affirmative Action/Equal Opportunity Employer LIBRARY Michigan State University 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 5/08 K:IProlecc&Pres/ClRC/DateDue.indd ROLE OF THE SPHINGOMYELIN/CERAMIDE PATHWAY IN DIABETIC RETINOPATHY By Madalina Opreanu A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Microbiology and Molecular Genetics 2010 ABSTRACT ROLE OF THE SPHINGOMYELIN/CERAMIDE PATHWAY IN DIABETIC RETINOPATHY By Madalina Opreanu I Diabetic retinopathy (DR) stands as a sight threatening disease without effective therapeutic options. Early DR is recognized to be a persistent low-grade chronic inflammatory disease. Acid (ASMase) and neutral (NSMase) sphingomyelinases (SMases) are important early responders in inflammatory cytokine signaling and key regulatory enzymes of sphingolipid metabolism. Sphingolipids are a major component of plasma membrane microdomains and sphingomyelin hydrolysis by SMases to bioactive lipid ceramide represents a prerequisite for inflammatory cytokines signaling. This study addresses the role of SMases in retinal vascular endothelium inflammation and development of microangiopathic lesions in the retinas affected by retinopathy. We first demonstrated that inhibition/gene silencing of both SMases decrease cytokine-induced cellular adhesion molecules expression in human retinal endothelial cells (HREC); yet a more pronounced anti-inflammatory effect was observed when ASMase inhibition/gene silencing was attained. Similarly, docosahexaenoic acid (DHA), the major (03 polyunsaturated fatty acid in the retina and well known anti-inflammatory agent, downregulated cytokine-induced inflammation in HREC. Interestingly, DHA decreased basal and cytokine-induced ASMase and NSMase expression and activity in HREC, further underlining the role of SMases as mediators of the inflammatory process. SMases pathway rather than ceramide de novo synthesis pathway was important for inflammatory signaling in HREC. To further demonstrate the role of SMase pathway in inflammation, the caveolae/lipid microdomains sphingolipid composition was then characterized. In support of SMases inhibition and displacement of ASMase from caveolae microdomains by DHA, we found a significant decrease in ceramide/sphingomyelin (Cer/SM) ratio in the caveolae fraction isolated from HREC treated with DHA; moreover, DHA prevented the TNFa-induced increase in the Cer/SM ratio in caveolar membrane microdomains and intracellular inflammatory signaling. To address the role of SMases in retinal inflammation and vessel loss in DR, an in vivo model of streptozotocin (STZ)-induced diabetes in rat was employed. ASMase, but not NSMase was upregulated in the diabetic retinas with an inflammatory status and microangiopathy lesions. DHA-enriched diet restored ASMase in diabetic retina to the control levels and prevented retinal inflammation and vessel loss. More importantly, DHA supplementation specifically prevented vascular ASMase upregulation in the retinas of a rat model with vasodegenerative phase of DR. To directly investigate the role of ASMase in development of retinal acellular capillaries and ocular neovascularization, we next used accelerated models of retinopathy by inducing ischemia/ reperfusion (I/R) injury and oxygen-induced retinopathy (OIR) in wild type (ASMase+/+) and ASMaseT mouse retina. ASMase'f mouse retina was significantly protected against retinal inflammation, vascular degeneration and ocular neovascularization. In summary, this is the first study to show that ASMase, a key regulatory enzyme of sphingolipid metabolism, is a novel and fundamental mediator and a promising therapeutic target for the prevention of retinal vascular inflammation and diabetic retinopathy. ACKNOWLEDGEMENTS I would like to deeply thank my committee members Dr. Julia V. Busik, Dr. Walter J Esselman, Dr. Andrea Amalfitano, Dr. Michele F luck, Dr. Karl L. Olson and Dr. Richard C. Schwartz for their valuable guidance and support for all these years. Especially I would like to dedicate my special gatitude to my mentor Dr. Julia V. Busik who guided my steps from the very early days of my research journey, teching me with enthusiasm and passion special techniques, opening my "mind eyes" towards a critical thinking; nevertheless, I would like to express my deep appreciation for her frendship, insight and understanding. I also want to express my profound gratitude for my mentor Dr. Walter J. Esselman who always offered me with the most kind and warmest support and encouragement so I can manage easier the uphills and downhills of my graduate career. Many thanks also go to Dr. Karl L. Olson and Dr. Narayanan Paraeswaran for their unconditional opening of their labs for me to apply any resources and for their insightful suggestions on experimental and technical apporaches. And more, Dr. Gloria Ines Perez in the Department of Physiology who contributes tremendously to my research by providing me the tools necessary for my experiments and who's door was always opened for help, advice and technical support. I am also very thanka for the fiiendship and valuable help of all my lab mates, Svetlana Bozack, Todd A. Lydic, Maria Tickhonenko, Kelly M. McSorley, Andrew Sochacki, Matt Faber in Dr. Busik lab. Particularly I thank Svetlana Bozack for her unlimited friendship and technical support; and Todd Lydic for his great assistance with iv mass spectrometry analysis. I would also like to thank my other lab fellows from Dr. Olson Lab and Dr. Parameswaran Lab, especially Chris Green and Sonika Patial for their valuable discussions and friendship. At last, but not least, I express my deep appreciation for my husband, for my parents and my entire family for their unflagging love and tremendous support during my graduate career. TABLE OF CONTENTS LIST OF TABLES x LIST OF FIGURES , xi LIST OF ABBREVIATIONS xiv 1. LITERATURE REVIEW 1 1. Diabetic Retinopathy 1 1.1. Overview ............................................................................................................... l 1.2. Histopathological and clinical manifestations ....................................................... 2 1.3. Epidemiology ........................................................................................................ 4 2. Dyslipidemia and diabetic retinopathy 6 2.1. Insulin and lipid metabolism ................................................................................. 6 2.2. Diabetic dyslipidemia ............................................................................................ 7 2.3. Retinal fatty acid profile and diabetic retinopathy .............................................. 11 3. Inflammatory mechanisms in diabetic retinopathy 17 3.1. Adhesion molecules in diabetic retinopathy ........................................................ 17 3.2. Proinflammatory cytokines in diabetic retinopathy ............................................ l8 4. Sphingomyelinase and Inflammation 23 4.1. Neutral sphingomyelinase ................................................................................... 23 4.2. Acid sphingomyelinase ....................................................................................... 25 4.3. Sphingomyelinases, plasma membrane microdomains and inflammatory signal transduction ................................................................................................................ 30 5. Retinal vascular endothelium 37 5.1. Overview ............................................................................................................. 37 5.2. Sphingomyelinases and vascular endothelium .................................................... 38 6. Hypothesis and objective of thesis 40 7. References 44 II. DOCOSAHEXAENOIC ACID INHIBITS CYTOKINE SIGNALING BY DOWNREGULATING SPHINGOMYELINASES IN HUMAN RETINAL ENDOTHELIAL CELLS 65 1. Abstract 65 2. Introduction 67 3. Materials and Methods 70 3.1. Reagents and supplies .......................................................................................... 70 3.2. Cell culture .......................................................................................................... 71 3.3. Fatty acid treatment ............................................................................................. 71 3.4. Sphingomyelinase assay ...................................................................................... 72 3.5. MCD and cholesterol treatment ........................................................................... 72 3.6. Cholesterol measurement .................................................................................... 73 3.7. Western blot analysis ........................................................................................... 73 3.8. Gene silencing of ASMase and NSMase ............................................................. 74 vi 3.9. Quantitative real-time polymerase chain reaction ............................................... 74 3.10. Isolation of caveolae/lipid rafts membrane domains ......................................... 75 3.11. Isolation of plasma membrane domains ............................................................ 76 3.12. Lipid extraction ................................................................................................. 76 3.13. Mass spectrometry analysis ............................................................................... 76 3.14. Statistical Analysis ............................................................................................ 78 4. Results 79 4.1. Dose response of TNFa and IL-IB induced cellular adhesion molecules (CAMS) expression in HREC ................................................................................................... 79 4.2. TNFa and IL-lB induce ASMase and NSMase activation in HREC .................. 79 4.3. Inhibition of cytokine-induced ASMase and NSMase activation by DHA in HREC ......................................................................................................................... 80 4.4. Effect of ASMase and NSMase inhibition and/or gene silencing on TNFa and IL-IB induced CAMS expression ................................................................................ 81 4.5. Inhibition of ASMase and NSMase activity by DHA ......................................... 82 4.6. Inhibition of ASMase and NSMase activity by MCD ......................................... 82 4.7. No effect of ceramide de novo synthesis inhibition on TNFa and IL-IB inducedCAMs expression in HREC ........................................................................... 84 5. Discussion 106 6. References 112 III. DOCOSAHEXAENOIC ACID REGULATES CAVEOLAR ACID SPHINGOMYELINASE EXPRESSION AND PROMOTES SPHINGOMYELIN ENRICHEMENT OF MEMBRANE MICRODOMAINS IN HUMAN RETINAL ENDOTHELIAL CELLS 119 1. Abstract 1 l9 2. Introduction 121 3. Materials and Methods 124 3.1. Reagents and antibodies .................................................................................... 124 3.2. Cell culture ........................................................................................................ 124 3.3. Fatty acid treatment ........................................................................................... 125 3.4. Sphingomyelinase assay .................................................................................... 125 3.5. Western blot analysis ......................................................................................... 125 3.6. Gene silencing of ASMase ................................................................................ 125 3.7. Isolation of caveolae/lipid rafts membrane microdomains ............................... 126 3.8. Isolation of plasma membrane domains ............................................................ 127 3.9. Lipid extraction ................................................................................................. 127 3.10. Mass spectrometry analysis ............................................................................. 128 3.11. Irnmunofluorescence assay .............................................................................. 129 3.12. Statistical analysis ........................................................................................... 130 4. Results 131 4.1. ASMase expression and activity in human retina] cells .................................... 131 4.2. ASMase cellular distribution in human retinal cells ......................................... 131 4.3. DHA effect on ASMase activity in human retinal cells .................................... 132 4.4. Downregulation of caveolar ASMase by DHA ................................................. 132 vii 4.5. Sphingomyelin enrichment of caveolae/lipid rafts by DHA ............................. 133 4.6. Gene silencing of ASMase prevents pro-inflammatory cytokine- induced HREC activation .................................................................................................................. 134 5. Discussion 148 6. References 152 IV. DOCOSAHEXAENOIC ACID SUPPLEMENTATION PREVENTS UPREGULATION OF PROIN F LAMMATORY/ ANGIOGENIC MOLECULES AND VESSEL LOSS IN DIABETIC RETINA THROUGH DOWNREGULATION OF ACID SPHINGOMYELINASE 158 1. Abstract 158 2. Introduction 160 3. Materials and Methods 163 3.1. Reagents and antibodies .................................................................................... 163 3.2. Rat model of type 1 diabetic retinopathy .......................................................... 163 3.3. The experimental diet ........................................................................................ 164 3.4. Isolation of retinal vasculature .......................................................................... 164 3.5. Histological assessement of retinal capillaries .................................................. 165 3.6. Simultanoeus extraction of proteins and RNA .................................................. 165 3.7. Quantitative real-time polymerase chain reaction ............................................. 166 3.8. Western blot ....................................................................................................... 167 3.9. Irnmunofluorescence assay ................................................................................ 167 3.10. Rat model of retinal ischemia-reperfusion ...................................................... 167 3.11. Slide preparation and quantification of ASMase staining of the blood vesselsl68 3.12. Statistical analysis ........................................................................................... 169 4. Results 170 4.1. DHA enriched diet prevents the upregulation of retinal ASMase in one month diabetic rat model .................................................................................................. 170 4.2. DHA enriched diet prevents the upregulation of retinal proinflammatory/angiogenic mediators in one month diabetic rat model ................ 171 4.3. DHA enriched diet prevents the upregulation of retinal ASMase in nine month diabetic rat model ................................................................................................... 171 4.4. DHA enriched diet prevents the upregulation of retinal proinflammatory/ angiogenic mediators in nine month diabetic rat model ........................................... 172 4.5. DHA enriched diet prevents retinal vessel loss in nine month diabetic rat model ........................................................................................................................ 172 4.6. Upregulation of ASMase and proinflammatory cytokines TNFa and IL-IB in human diabetic retina ............................................................................................... 173 4.7. ASMase expression in the rat retinal vasculature .............................................. 173 4.8. DHA supplementation prevents the upregulation of vascular ASMase in the rat retina injured by I/R .................................................................................................. 174 5. Discussion 191 6. References 195 V. ACID SPHINGOMYELINASE GENETIC DEFICIENCY PREVENTS UPREGULATION OF INFLAMMATORY/ANGIOGENIC MEDIATORS, viii VESSEL LOSS AND PATHOLOGICAL NEOVASCULARIZATION IN THE RETINA WITH MICROANGIOPATHY LESIONS 201 1. Abstract 201 2. Introduction 202 3. Materials and Methods 206 3.1 . Reagents ............................................................................................................ 206 3.2. Genotyping transgenic mice by PCR and Southemblot .................................... 206 3.3. Mice ................................................................................................................... 208 3.4. Isolation of retinal vasculature .......................................................................... 209 3.5. Histological assessement of retinal capillaries .................................................. 209 3.6. Simultanoeus extraction of proteins and RNA .................................................. 210 3.7. Quantitative real-time polymerase chain reaction ............................................. 210 3.8. Statistical analysis ............................................................................................. 210 4. Results 211 4.1. Retinal I/R induces upregulation of ASMase gene expression in the ASMase +/+ retina ......................................................................................................................... 211 4. 2. ASMase genetic deficiency prevents upregulation of cellular adhesion molcules in the I/R injured retina ............................................................................................. 211 4. 3. ASMase genetic deficiency prevents upregulation of inflammatory/ angiogenic mediators 1n the I/R injured retina ............................................................................ 212 4.4. ASMase genetic deficiency prevents the development of acellular capillaries in the I/R injured retina ................................................................................................. 212 4.5. ASMase genetic deficiency prevents ocular neovascularization in OIR mouse model ........................................................................................................................ 213 5. Discussion 221 6. References 226 ix LIST OF TABLES Table 4.1. Body weight gain and blood glucose of experimental animals with one of month diabetes .......................................................................................................... 175 Table 4.2. Body weight at the end of the study, blood glucose and HbAl c of experimenatal animals with nine months of diabetes ............................................... 175 Table 4.3. Fatty acid composition of the control diet and DHA (fish oil)- enriched diet ............................................................................................................................ 176 Table 4.4. RT-PCR primers for rat ........................................................................... 177 Table 4.5. RT-PCR primers for human .................................................................... 177 Table 5.1 RT-PCR primers for mouse ...................................................................... 214 LIST OF FIGURES Images in this thesis/dissertation are presented in color Fig. 1.1. Fatty acids classification .............................................................................. 14 Fig. 1.2. Sphingolipids chemical structures ................................................................ 15 Fig. 1.3. Cerarnide generation by Sphingomyelinase-mediated sphingomyelin hydrolysis ................................................................................................................... 35 Fig. 1.4. Receptor clustering and amplification of signal transduction by ceramide- enriched macrodomains .............................................................................................. 36 Fig. 1.5. Diagram of working hypothesis ................................................................... 43 Fig. 2.1. Dose response of cytokine-induced CAMS expression in HREC ................ 85 Fig. 2.2. Cytokine induced ASMase and NSMase activation in HREC ..................... 86 Fig. 2.3. Inhibition of cytokine induced ASMase and NSMase activation, as well as ASMase and NSMase expression by DHA in HREC ................................................ 88 Fig. 2.4. Inhibition of ASMase and NSMase cytokine-induced activation by DHA and sphingomyelinases inhibitors ............................................................................... 90 Fig. 2.5. Decrease in TNF a induced CAMS expression by ASMase and NSMase inhibition in HREC ................................................................................................. 91 Fig. 2.6. Inhibition in cytokine-induced CAMS expression by ASMase and NSMase gene Silencing in HREC ............................................................................................. 93 Fig. 2.7. Inhibition of ASMase and NSMase activity by DHA .................................. 97 Fig. 2.8. No direct effect of DHA on sphingomyelinase (SMase) activity in a cell- free system .................................................................................................................. 98 Fig. 2.9. Effect of cholesterol depletion on ASMase and NSMase activity in HREC ......................................................................................................................... 99 Fig. 2.10. No effect of de novo ceramide production pathway inhibition on cytokine induced CAMS expression in HREC ........................................................................ 102 Fig. 3.1. ASMase expression in human retinal cells ................................................ 135 Fig. 3.2. ASMase cellular distribution in human retinal cells .................................. 136 xi Fig. 3.3. The effect of DHA on ASMase activity in HRPE and HMC .................... 139 Fig. 3.4. Downregulation of caveolar ASMase by DHA ..................................... 140 Fig. 3.5. The effect of DHA on sphingomyelin and ceramide content of caveolae membrane microdomain in HREC ........................................................................... 141 Fig. 3.6. The effect of DHA and ASMase gene Silencing on IL-1 B-induced HREC activation .................................................................................................................. 146 Fig. 4.1. The effect of DHA-enriched diet on ASMase retinal gene expression in rats with one month of diabetes ....................................................................................... 178 Fig. 4.2. The effect of DHA-enriched diet on retinal inflammatory/angiogenic molecules gene expression in rats with one month of diabetes ................................ 179 Fig. 4.3. The effect of DHA-enriched diet on retinal ASMase and VEGF protein expression in rats with one month of diabetes .......................................................... 180 Fig. 4.4. The effect of DHA-enriched diet on retinal ASMase gene expression in rats with nine months of diabetes .................................................................................... 181 Fig. 4.5. The effect of DHA-enriched diet on retinal ASMase protein level in rats with nine months of diabetes .................................................................................... 182 Fig. 4.6. The effect of DHA-enriched diet on retinal inflammatory/angiogenic molecules gene expression in rats with nine months of diabetes ............................. 183 Fig. 4.7. The effect of DHA-enriched diet on retinal vessel loss in rats with nine months of diabetes ................................................................................................ 184 Fig. 4.8. Changes in retinal gene expression in control and diabetic human retina ......................................................................................................................... 186 Fig. 4.9. Rat retinal ASMase expression ................................................................. 187 Fig. 4.10. Retinal vascular ASMase expression after retinal I/R injury in rats ....... 188 Fig. 5.1. Retinal gene expression of ASMase in the l/R retinopathy model ........... 215 Fig. 5.2. Retinal gene expression of NSMase in the I/R retinopathy model ........... 216 Fig. 5.3. Retinal gene expression of cellular adhesion molecules in the I/R retinopathy model ..................................................................................................... 217 xii Fig. 5.4. Retinal gene expression of inflammatory/angiogenic molecules in the I/R retinopathy model .................................................................................................. 218 Fig. 5.5. Retinal capillary degeneration in retinal ischemia/reperfusion in I/R retinopathy mouse model ......................................................................................... 219 Fig. 5.6. Retinal neovascularization in oxygen-induced retinopathy in mice .......... 222 xiii AGE ASMase BSA CAM Cer COX2 CRD DD DHA DNA DR Elovl FADD FAN HDL HMC HREC HRPE HSL ICAM-l IL I/R LIST OF ABBREVIATIONS advanced glycation end product acid sphingomyelinase bovine serum albumin cell adhesion molecule ceramide cyclooxigenase 2 caveolae-related domains death domaain docosahexaenoic acid (22:6n3) deoxyribonucleic acid diabetic retinopathy elongase FaS-associating protein with DD factor associated with NSMase activation high density lipoprotein human Muller cells human retinal endothelial cells human retinal pigment epithelial cells hormone—sensitive lipase intercellular cell adhesion molecule -1 interleukin ischemia/reperfusion xiv LAMP 1 LDL LPL MCD MMP MS MUFA NSMase NF-KB OIR PKC PCR RNA PUFA ROS SM SMase STZ TRADD VCAM-I VEGF VLDL lysosomal associated protein 1 low density lipoprotein lipoprotein lipase methyl-B-cyclodextrin matrix metalloprotease mass Spectrometry monounsaturated fatty acid Neutral sphingomyelinase nuclear-factor kappa B oxygen-induced retinopathy protein kinase C polymerase chain reaction ribonucleic acid polyunsaturated fatty acid reactive oxygen species sphingomyelin Sphingomyelinase streptozotocin tumor necrosis factor TNF receptor associated DD vascular cell adhesion molecule-1 vascular endothelial growth factor very low density lipoprotein XV Chapter I Literature Review 1. Diabetic Retinopathy 1.1. Overview Current estimates suggest that approximately 135 million individuals across the world have diabetes mellitus (1). In the US, over 23.6 million people are affected by diabetes with Significant morbidity and mortality (2). Diabetic retinopathy (DR) stands as the most common and disabling microvascular complication of diabetes and results in blindness for over 12,000 people with diabetes each year (2). Moreover, DR is the primary cause for blindness in the working age population worldwide (3) and in 2004, vision loss from DR accounted for approximately $500 million in direct medical costs among Americans age 40 or older (4). To place this in perspective, it is estimated that the number of Americans 40 years or older with DR will triple during the period of time from 2005 to 2050, from 5.5 million in 2005 to 16.0 million in 2050 (5). In conclusion, it is largely recognized that visual impairment among people with I diabetic retinopathy is a major disability with a profound impact on patient independence and can result in depression, reduced mobility and overall, poor quality of life (6). Consequently, diabetic eye disease continues to represent an important socio-economic burden on society and remains a challenge for the ophthalmology services provision. 1.2. Histopathological and Clinical Manifestations Diabetic retinopathy represents the ocular expression of diabetic microvascular complications. The natural history of the DR has been classified into an early, non- proliferative stage (background retinopathy) and a late, proliferative stage. The early, non-proliferative stage of DR has distinct vascular lesions that are histologically characterized by the existence of saccular capillary microaneurysms, retinal hemorrhage, venous dilation and beading, retinal lipid exudates, pericyte degeneration, occlusion and degeneration of retinal capillaries (7-8). The mechanisms considered to be involved in diabetic retinal vascular lesions include obstruction of the vascular lumen by leukocytes or platelets; death of endothelial cells due to pathophysiological processes within the vascular cells themselves; or endothelial cell death secondary to products generated by other cells in the retina that are in close contact with these vascular cells (such as neurons or glial cells) (7). During the early phase of DR, clinical signs include retinal microaneurysms and dot hemorrhages that are present in almost all patients with type 1 diabetes of a duration 20 years (9) and in approximate 80% of those affected by type 2 diabetes for the same duration of time (10). As the disease progresses, the number and size of intraretinal hemorrhages increases, and is accompanied by the appearance of cotton-wool spots on the retinal background, indicative of ischemia due to local failure of the retinal microvascular circulation. During the ophthalomoscopic investigation of the retina, retinal veins appear dilated, tortuous and with irregular caliber, whereas arteries may appear white; fluorescein angiography may reveal non-perfiised retinal arteries (11). The late, proliferative stage of DR is histologically characterized by the presence of aberrant new vessels, and fibrous tissue proliferation on the retinal surface that can extend into the vitreous. The newly synthesized blood vessels are abnormal and fragile. They tend to leak blood into the center of the eye inducing blurred vision and/or causing blindness. As DR progresses, macular edema develops in the retina (due to the breakdown of the blood-retinal barrier) resulting in leakage of plasma from the small retinal vessels into the maculaand leading to severe loss of central vision (11). The clinical signs of late stage DR are due to the proliferating retinal vessels along with the glial tissue that anchor into the vitreous and lead to traction of the retina (3). In the most severe cases, the fibrovascular tissue proliferation leads to total retinal detachment and vitreous hemorrhage (12). Retinal neovascularization most commonly takes place along the temporal vascular arcades and the optic nerve head. Retinal neovascularization is an important contributor to the visual impairment and blindness in diabetes. Current approaches to prevention of DR involves general medical measures such as control of blood glucose, blood pressure and serum lipids (11), whereas treatment of DR involves specific therapeutic strategies that target retinal abnormalities encountered during the course of the disease. The surgical options (laser panretinal photocoagulation and vitreo-retinal surgical interventions) applied locally at the retinal level are highly invasive, traumatic to the retinal tissue and do not prevent visual impairment. Although the medical and surgical management of diabetes represent the mainstay of DR management, they are not fiilly protective against visual impairment and patients suffering blindness from diabetes are still arising, with approximately 8,000 new cases reported annually. Therefore, new therapeutic approaches based on the etiology of early diabetic microvascular lesions need to be developed and evaluated in clinical trials to ultimately improve the outcome for patients with DR. 1.3. Epidemiology Wisconsin Epidemiological Study of Diabetic Retinopathy (WESDR) was the most comprehensive study of retinal neovascularization within a diabetic population, where the prevalence and incidence of DR was investigated in more than 2000 patients with diabetes (9-10, 13-14). This study showed that the prevalence of all retinopathy was approximately 17% in patients with diabetes for less than 5 years, and 97.5% in patients with diabetes for more than 15 years. The increase in prevalence of proliferative retinopathy was strongly correlated with greater duration of diabetes and it varied from 1.2% in patients with diabetes mellitus for less than 10 years to more than 67% in patients with diabetes for 35 years or more (9). This study on DR concluded that there is a strong association between the frequency and severity of retinopathy and the duration of diabetes. Moreover, the severity of DR was also related to a diagnosis at a younger age, increased levels of glycosylated hemoglobin, the use of insulin, higher systolic blood pressure, proteinuria and a small body mass (10). New clinical data identified dyslipidemia as a critical factor in the development of diabetic retinopathy. The Diabetes Control and Complication Trial (DCCT)/Epidemiology of Diabetes Interventions and Complications (EDIC) identified a strong association between the severity of DR in type 1 diabetes and lipid metabolism (15). This study was conducted on patients with type 1diabetes and revealed a strong association between severity of retinopathy in type 1 diabetes and the size of the particles of three major classes of serum lipoproteins, very low density, low density and high density lipoprotein (VLDL, LDL and HDL) as well as LDL concentration. Cross- sectional studies Showed positive associations between the severity of retinopathy and total- and LDL-cholesterol levels and the LDL-HDL cholesterol ratio (16). The Early Treatment Diabetic Retinopathy Study has demonstrated that higher levels of serum lipids are associated with an increased risk of development of hard exudates in the macula and visual loss (17-18). Several studies Showed that lipid-lowering dietary (19) and drug (20) therapy may lead to regression of retinal hard exudates, and that a diet high in polyunsaturated fatty acids may protect against retinopathy (21). 2. Dyslipidemia and Diabetic Retinopathy Dyslipidemia is not only a major metabolic disorder of diabetes mellitus, but also a critical factor in the development of DR. Diabetic dyslipidemia with lipoprotein abnormalities become manifested during the asymptomatic diabetic prodrome and represent a high risk for vascular complications (22). 2.1. Insulin and Lipid Metabolism Insulin, a polypeptide hormone secreted by B cells of the islets of Langerhans in the pancreas, exerts an essential metabolic regulation not only on carbohydrate metabolism, but also on lipid metabolism and protein synthesis. Insulin decreases triacylglycerol degradation and hence the level of circulating fatty acids by inhibiting hormone-sensitive lipase (HSL) activity in the adipose tissue (23). Moreover, insulin increases glucose transport and metabolism in adipocytes and provides the substrate glycerol 3-phosphate for triacylglycerol synthesis. In adipose tissue and muscle, insulin also enhances the activity of lipoprotein lipase (LPL) and thus provides fatty acids for esterification (24). LPL is then transported to the surface of endothelial cells where it hydrolyzes the lipid chore of chylomicrons and low/very low density lipoprotein particles and thus facilitate in the removal of triglyceride-rich lipoproteins from the blood stream. Patients with either type 1 or type 2 diabetes have low adipose LPL activity, a higher rate of HDL-metabolism and increase in plasma triglycerides level (25). Fatty acid synthesis and remodeling is also under insulin control. Fatty acids are classified according to their structure by the number of carbons, double bonds and proximity of the first double bond to the methyl (-CH3) end of the fatty acyl chain. According to the number of the double bonds, fatty acids are classified as saturated (no double bonds), mono-unsaturated (MUFA, one double bond) and poly-unsaturated (PUFA, multiple double bonds) and are presented in Figure 1.1. Saturated, mono- unsaturated and poly-unsaturated fatty acids are synthesized from dietary precursors (glucose, 16:0, 18:1n9, 18:2n6, 18:2n3, 18:2n6 and 20:5n3) through a series of elongation (Elovl-2,—4,-5 and -6) and desaturation (AS—desaturase, A6-desaturase and A9-desaturase) reactions. Insulin is the most potent activator of A5, A6 and A9 desaturase, where A5 and A6 desaturases represent rate-limiting enzymes in fatty acid metabolism (25-30). Therefore, low insulin availability or insulin resistance will result in impaired desaturase expression, leading to substrate accumulation and product depletion. The major impact is observed on long chain polyunsaturated a) 3 fatty acids that are no longer synthesized in the desirable amounts. The lack of insulin in type 1 diabetes and insulin resistance in type 2 diabetes will promote a fatty acid profile with high saturation index and Short fatty acyl chain and will lead to a Significant increase in HSL activity and decrease in PLP activity, with a profound impact on plasma lipid levels, lipid profile and fatty acid composition. 2.2. Diabetic Dyslipidemia 2.2.1. Lipoproteins and Triglycerides The natural course of development of type 2 diabetes (or insulin-resistant diabetes) affects virtually all lipids and lipoproteins (31-32). The metabolic milieu is characterized by accumulation of chylomicrons and VLDL remnants, triglyceride enrichment of HDL and LDL particles, resulting in decrease level of HDL particles and formation and accrual of small, dense LDL particles (33). Increased triglyceride production along with decreased catabolism of triglyceride-enriched lipoproteins heightens hypertriglyceridemia. Studies have shown that LDL particles become small and dense in approximatively 90% of the patients whom triglycerides levels are > 200 mg/dL; when triglyceride levels are shifted 90 mg/dL, almost all LDL lipoprotein particles are of the larger, buoyant range (34). The epidemiological data on the prevalence of dyslipidemia and phenotype distribution showed that, regardless of a poor glycemic control, total triglyceride and cholesterol levels are the same when compared patients with insulin-dependent type 1 diabetes and nondiabetic, control, healthy subjects age matched (35). However, increased LDL-cholesterol levels and decrease in HDL-cholesterol have been found in diabetic group compared to nondiabetic group (3 5). More importantly, the lipid abnormalities may improve with a good glycemic control, except for HDL-cholesterol levels (35). Kordonouri et al. also Showed that HDL-cholesterol is the most important variable, tightly correlated to the development of retinal lesions in children with type 1 diabetes (36). 2.2.2. Long chain PUFAS Over the past few years, there has been an increasing interest in exploring the role of long chain PUFAS in pathophysiology of diabetic metabolic complications. These PUFAS contains more than one cis double bond and are classified as (06 or (133-PUFAS according to the location of the first double bond compared to the fatty acid methyl end as shown in Figure 1.1. (03- PUFAS are regarded as important anti-inflammatory mediators (37-39), whereas (06- PUFAS are considered to have pro-inflammatory properties (40). More importantly, diabetic dyslipidemia is characterized by an elevated (06 to w3-PUFA acid ratio (41-42) that can "incline the body physiologic equilibrium" toward an inflammatory state. Ferruci et al. studied the relationship between plasma PUFAS and circulating inflammatory markers in a community based-sample and found that increased total (0-3 PUFAS levels were associated with lower levels of inflammatory markers (IL-6, IL-lra, TNFa, C reactive protein) and higher levels of anti-inflammatory markers (soluble IL-6r, IL-lO) (43-44). The authors concluded that (D3-PUFAS are beneficial in patients affected by diseases with an active inflammatory component. 2.2.3. Sphingolipids New research revealed the importance of another class of lipids that are dysregulated in diabetes, the sphingolipids. Sphingolipids represent a complex class of lipids and the prefix “sphingo-” that appears in the nomenclature of this lipid class was first attributed by J.L.W. Thudichum in 1884 because the enigmatic nature of these molecules reminded him of the riddle of the Sphinx. In 1974, Herbert Carter and colleagues introduced the “sphingolipide” name. Sphingolipids are extremely versatile molecules. Relative to phospholipids, sphingolipids are enriched in saturated long fatty acyl chains, are more hydrophobic, have a higher melting temperature and can undergo a tighter packing in the cellular membrane due to the lack of unsaturated acyl chains with kinked structures (Figure 1.2) (45-46). Sphingolipids have a chemical structure composed of a 1,3-dihydroxy—2-aminoalkane backbone, also known as the sphingoid base. Sphingosine, the most prevalent backbone of mammalian sphingolipids is characterized by a (28,3R,4E)-2-amino-4-octadecene-1,3-diol chemical structure. Saturation of the sphingosine backbone results in sphinganine, which represents another sphingoid base frequently used to form mammalian sphingolipids. Attachment of a fatty acid to the sphingosine backbone via an amide bond results in formation of ceramides; attachment of a variety of polar headgroups to the ceramide structure results in formation of complex sphingolipids, such as Sphingomyelin, gangliosides, sulfatides, globosides or cerebrosides. Sphingolipids such as ceramide and glucosylcerarnide, while representing a relatively minor component of the lipid milieu in most tissues, may be among the most pathogenic lipids in the onset of diabetic metabolic complications, including insulin resistance, pancreatic B-cell failure and vascular dysfunction (47-48). Circulating factors associated with diabetes (e. g. saturated free fatty acids, inflammatory cytokines) selectively induce enzymes that promote ceramide synthesis and stimulate the accumulation of ceramide and various ceramide metabolites (49-52); these sphingolipids have been Shown to amass in tissues from rodents (53-54) and humans (55-56) with type 2 diabetes. Extensive lipidomic profiling revealed ceramide accumulation in the muscle tissue from rats on a high fat diet for three weeks or from streptozotocin diabetic rats, one month after diabetes induction (54). Related to these findings, Straczkowski et al. have shown elevated ceramide levels in the skeletal muscle of obese men at risk of developing type 2 diabetes (57). Interestingly, a fish oil (DHA) enriched diet significantly reduced ceramide content in the mouse muscle tissue (58). Dysregulation of sphingolipid metabolism in diabetes emerges as an important pathway for development of the deleterious clinical manifestations associated with this 10 disease. Importantly, sphingolipid metabolism was found to be Significantly altered in diabetic retina (59). Therefore, pharmacologic inhibition or genetic ablation of key regulatory enzymes of sphingolipid metabolism may identify new therapeutic targets for combating insulin resistance, B-cell failure and vascular dysfunction of diabetic metabolic disease. 2.3. Retinal Fatty Acid Profile and Diabetic Retinopathy The normal retina has a very specific fatty acid profile, with the highest level of long-chain PUFAS in the body. Various studies have identified DHA, an (0-3 PUFA as being enriched in the retinal tissue (60-61). Work from our group demonstrated that DHA represents 45.37 d: 1.32 mole % of total fatty acids identified in the retina (61). Omega 3- PUFAS and particularly DHA have the ability to modulate various biological processes involved in retinal vascular inflammation, retinal capillary structure and integrity alterations, and retinal neovascularization (62). Therefore, (03 long-chain PUFAS and mainly DHA status in the retina is closely related to normal retinal structure and function (62). Long chain PUF A synthesis and remodeling involves a series of desaturation (A5- desaturase, A6-desaturase, A9-desaturase) and elongation (Elov11-7) reactions. The elongases Elov12 and Elov16 are ubiquitously present in most tissues; yet, the retinal tissue express very high levels of Elov12 and this elongase is involved in several steps of DHA synthesis (61, 63). Elovl4 has a restricted tissue specificity, being highly expressed in the retina (64-66), thymus and skin (65), and to a lesser extent in the brain (65-66) and testis (66). Insulin deficiency or complete absence of insulin downregulates Elov12 and 11 Elovl4 elongases Specifically in the retina, and thus precludes the synthesis of various retinal PUFAS, and particularly (D3-PUFAS and DHA (61). We have found that retinas isolated from type 1 diabetic animals had a 28% reduction in DHA levels when compared to control (61). Previous studies have also demonstrated decreased levels of oo3-PUFAS, and mainly DHA in plasma of diabetic children (67), as well as in human retina of diabetic eyes (68). More importantly, decreased retinal DHA levels, and an increase in the w6-to- (o3 PUFA ratio in diabetic retina was associated with significant upregulation of [CAM-l and inflammatory cytokine (VEGF, TNFa and IL-6) gene expression, suggesting that DHA tissue insufficiency may create pro-inflammatory conditions in the retina, potentially contributing to the development of DR (61). Our study also demonstrates that retinal fatty acid metabolism is distinct from the metabolic changes that occur in diabetic plasma or diabetic liver (61 ). Several studies have identified DHA retinal insufficiency to be associated with alterations in retinal function (reviewed in (62)) and DHA deficiency has also been documented to be associated with certain retinal degenerative diseases, such as retinitis pigrnentosa, retinopathy of prematurity and age-related macular degeneration (69-73). Taken together, all these demonstrate that DHA has a critical role in maintaining the normal structure and function of the retina. A study by Connor et al. showed a beneficial effect of dietary DHA in reducing pathological retinal angiogenesis and thus preventing the development of retinopathy (74) in an oxygen-induced retinopathy (retinopathy of prematurity) animal model. Furthermore, it has been Shown that DHA supplementation could ameliorate visual processing deficits (62). In vitro studies from our group demonstrated a significant 12 protective effect of DHA against cytokine induced inflammation in human retinal endothelial cells (39-40). However, the protective effect of DHA against diabetic retinal inflammation and microangiopathic lesions has not been addressed and represents an objective of my study. 13 Saturated Fatty Acids 0 Palmitic Acid (16:0) HOW/WC”: Mono Unsaturated Fatty Acids HO _ WWW. Oleic Acid (18:1,n9) O Poly Unsaturated Fatty Acids HO N6 Linoleic Acid (18:2,n6) (n6-PUFA) OWMH3 Poly Unsaturated Fatty Acids COOH Docosahexaenoic Acid CH (DHA, 22:6,n3) (n3-PUFA) N3 3 Figure 1.1. Fatty acids classification. Fatty acids classification is based on the number of carbons in the chain length, the number of double bonds and the localization of the first double bond related to the methyl end. Based on these considerations, fatty acids can be classified as saturated, monounsaturated and polyunsaturated. 14 Sphingoid bases Sphingosine OH /_/I\/CH20H WM _ 5 NHz Dihydrosphingosine OH WNk/CHZOH NH2 Phytosphin osine 9 OH xc\\,#“\c/”\c//‘xa/“\sx”\cArbxg/Lxc/CHon OH NHg OH Dehydrophytosphingosine /\/\/\/\/—/\/\/i\/ CHZOH OH NHg Figure 1.2. Sphingolipids chemical structures. Sphingoid bases are long-chain aliphatic amines, with two or three hydroxyl groups, and ofien a characteristic trans-double bond in position 4; they are 2-amino-1,3-dihydroxy-alkanes or alkenes with (2S,3R)-erythro stereochemistry, with various structural modifications. 15 Ceramide Sphingomyelin Figure 1.2 (continued). Sphingolipids chemical structures. Ceramide consists of a sphingoid base linked to a fatty acid via an amide bond. Sphingomyelin consists of a ceramide molecule with a phosphorylcholine moiety attached to position 1. 16 3. Inflammatory Mechanisms in Diabetic Retinopathy An extensive body of evidence, including both preclinical and clinical findings, supports the concept that very early stage diabetic retinopathy represents a persistent low- grade chronic inflammatory disease that involves increased expression of adhesion molecules on endothelial cell surfaces, eventuating in leukocyte recruitment, retinal cytokine production and activation of transcription factors that result in up-regulation of retinal inflammatory genes. These pathophysiological changes will be separately discussed and evidence to support their role in DR will be presented. 3.1. Adhesion Molecules in Diabetic Retinopathy The first line of evidence supporting a connection between inflammation and diabetic retinopathy came from an epidemiological data in 1964 Showing a decrease in severity of DR in diabetic patients treated with high doses of aspirin for rheumatoid arthritis (75). In a rat model of diabetic retinopathy, aspirin and meloxicam (nonsteroidal anti-inflammatory agents) prevented the development of early diabetic retinopathy, a result that correlated with suppress of intercellular adhesion molecule-1 (ICAM-1) expression and leukocyte adhesion (76). Adhesion molecules, especially ICAM-1 and vascular cell adhesion molecule-1 (VCAM-l), are involved in leukocyte attachment and transmigration into the vascular intima (77-78) that could result in early blood retinal barrier breakdown, capillary non-perfusion and endothelial cell injury and death. Evidence supporting this view are provided by findings of Significant increase in leukocyte density and retinal vascular ICAM-l expression in human eyes with DR (78). Moreover, a role of CD18/ICAM-l-mediated leukocyte adhesion in the pathogenesis of 17 early diabetes-induced leukostasis and blood-retinal barrier breakdown has been established. Reduced expression of ICAM-1 on endothelial cells or its leukocyte counter- receptor CD18 Significantly inhibits leukocyte adhesion, pericyte and endothelial cell loss and consequently the formation of acellular, degenerated capillaries in experimental models of diabetic retinopathy (79). Theoretically, an intervention at this level might carry an important role in designing future treatment strategies in DR. 3.2. Proinflammatory Cytokines in Diabetic Retinopathy Proinflammatory cytokines (tumor necrosis factor alpha (TNFa), interleukin 1 beta (IL-10), and vascular endothelial growth factor (VEGF) are increased in diabetic eyes and several inflammatory pathways are activated in the very early stages of DR (79- 82). TNFa is a proinflammatory cytokine implicated in a number of inflammatory diseases, including rheumatoid arthritis, Chron's disease, ankylosing spondylitis and psoriasis (83). In diabetes, TNFa has been involved in pancreatic B cell apoptosis (84) and insulin resistance (85), as well as in diabetic microvascular complications, including nephropathy (86) and diabetic retinopathy (76). TNFa acts through ligand-receptor interactions and initiate a number of inflammatory processes, such as upregulation of adhesion molecules expression, leukocyte recruitment and leukostasis, cellular apoptosis, chemoattraction of monocytes and plays a critical role in amplification of the immune reSponse through upregulation of the expression of numerous transcription factors, growth factors and other inflammatory mediators (86). Clinical and preclinical studies indicate an important role of TNFa in the pathogenesis of DR. Correlative clinical studies with 18 genetic data have implicated a TNFa polymorphism in confen'ing susceptibility to the disease (87). Moreover, retinal levels of TNFa are significantly increased in diabetic rats compared to control (76) and increased amounts of TNFa can be found in the extracellular matrix, vessel walls, endothelium and vitreous of diabetic eyes affected by proliferative retinopathy (80, 88-89). Playing a central role in the pathogenesis of DR, TNFa has been targeted with specific medication aimed to counteract its inflammatory effects. Systemic administration of etemacept, a soluble TNFa receptor/Fe fusion protein that acts as a competitive inhibitor to block the effects of TNFa on cells, resulted in reduction of the inflammatory process in DR, including leukocytes adhesion to the retinal vasculature, upregulation of ICAM-1, NF-KB activation and breakdown of the blood retinal barrier (76). The effects of etemacept were not evaluated on advanced histologic lesions in DR, but it was reported that mice genetically deficient in TNFa were protected against galactose-induced retinopathy (7). Administration of lnfliximab, a monoclonal antibody directed against TNFa, for two months in patients with advanced DR Showed positive responses, with improvement of visual acuity and reduction in macular edema and further improvements were seen after repeated infusions (90). Increased intraocular VEGF expression, a growth factor recently recognized to have cytokine properties, has been identified in rat and human diabetic retina (82, 91-94) and has become a focal point of current research on the pathogenesis of DR. The VEGFS are a family of peptides produced by alternative splicing fiom a single gene; VEGF 1'80me3 are particularly mitogenic for vascular endothelium and also amplify permeability at blood-tissue barriers (1 1). VEGF is synthesized by multiple cell types in 19 the retina, such as ganglion cells, Muller cells and pericytes (95-97). VEGF induces upregulation of lCAM-l expression in endothelial cells (98), resulting in chronic retinal leukostasis that plays a key role in endothelial cell injury and death and blood-retinal barrier breakdown (94). VEGF is a key molecule that plays a well-recognized role in promoting vascular permeability (99) and pathological ocular neovascularization (100). VEGF expression is mainly regulated by hypoxic stimulus; yet there is evidences that it also accumulates in the retina early in diabetes, before any retinal hypoxia is present (101 - 102). Therefore, VEGF upregulation is strongly involved in the pathogenesis of both non- proliferative (background) and proliferative DR (81-82, 94, 103). Specific inhibition of VEGF hinders ICAM-l upregulation, leukostasis, blood-retinal barrier breakdown and pathological ocular neovascularization in diabetic rats (98). Clinical trials using anti- VEGF therapeutic strategies intravitreously administered (pegaptanib, a VEGF inhibitor; ranibizumab and bevacizumab, monoclonal antibodies that bind all VEGF isoforms) showed promising results against advanced stages of DR (104-106). Among these, ranibizumab is the only anti-VEGF therapy that was approved by Food and Drug Administration for ophthalmologic use in neovascular (wet) age-related macular degeneration; yet, it's efficacy in DR remains to be investigated. Pro-inflammatory cytokine IL-lB levels are also increased in retinas from diabetic rats (107-108). In vivo studies with intravitreal injection of IL-IB or exposure of retinal endothelial cells to IL-lB in vitro was able to induce endothelial cell inflammation and death, resulting in degeneration of retinal vascular endothelium and formation of acellular capillaries (109). Caspase-l, the enzyme involved in production of proinflammatory cytokine IL-lB from pro-IL-l B, is activated in the retina of diabetic patients and diabetic 20 animal models (110). The involvement of IL-lB in pathogenesis of DR has been recently defined in diabetic mice in which caspase-1 was inhibited or the IL-IB receptor was deleted (108). Inhibition of caspase-1 using minocycline or inhibition of IL-lB signaling using IL-IB receptor genetic deficient mice protected the mice from development of retinal pathology (108). These data suggests a major role for IL-IB and its receptor in mediating signaling pathways responsible for endothelial cell activation and death, and consequent retinal vessel loss. Proinflammatory cytokines that are upregulated in diabetic retina (i.e. TNFa, IL- 10 and VEGF) are likely to activate nuclear factor-kappa B (NF-KB) pathway (40). NF- KB proteins consist of a family of structurally related transcription factors widely expressed in mammalian tissues, which control a large number of cellular processes, such as immune and inflammatory responses, developmental processes, cellular growth, and apoptosis (7). NF-IcB transcription factors represent a prerequisite for various gene expressions, including adhesion molecules, inflammatory cytokines, cyclooxygenase 2 (COX2) and matrix metalloproteases (MMPs) (111-112). These transcription factors are persistently active in various disease states, including chronic inflammation, arthritis, asthma, cancer, and heart disease (113). DR is hypothesized to be a chronic inflammatory disease and activation of NF-KB is well documented in the retinal vasculature of diabetic patients and animals (114-115). TNFa and IL-lB have been shown to activate cellular sphingomyelinases in various cell types (116-119). Stimulation of the sphingomyelin pathway by TNFa, andSphingomyelinases-induced membrane ceramide formation leads to activation of NF- KB and marked increase in nuclear NF -II ASMase upregulationd—DHA \/ Retinal vascular degenerationJ Diabetic retinopathyI Figure 1.5. Diagram of working hypothesis. Potential impact of decreased retinal (o3- PUFA DHA on retinal inflammatory status, ASMase upregulation, microvascular complications and progression of diabetic retinopathy. 43 7. References l. 10. ll. 12. Fong, D.S., Aiello, L.P., Fenis, F.L., 3rd, and Klein, R. 2004. Diabetic retinopathy. Diabetes Care 27:2540-2553. Varma, R. 2008. From a population to patients: the Wisconsin epidemiologic study of diabetic retinopathy. Ophthalmology 1 15: 1857-1858. Aiello, L.P., Cavallerano, J., and Bursell, SE. 1996. Diabetic eye disease. Endocrinol Metab Clin North Am 25:271-291. Rein, D.B., Zhang, R, Wirth, K.E., Lee, P.P., Hoerger, T.J., McCall, N., Klein, R., Tielsch, J.M., Vijan, S., and Saaddine, J. 2006. The economic burden of major adult visual disorders in the United States. Arch Ophthalmol 124:1754-1760. Saaddine, J.B., Honeycutt, A.A., Narayan, K.M., Zhang, X., Klein, R., and Boyle, LP. 2008. Projection of diabetic retinopathy and other major eye diseases among people with diabetes mellitus: United States, 2005-2050. Arch Ophthalmol 126:1740-1747. Sinclair, A.J., Bayer, A.J., Girling, A.J., and Woodhouse, K.W. 2000. Older adults, diabetes mellitus and visual acuity: a community-based case-control study. Age Ageing 29:335-339. Kern, TS 2007. Contributions of inflammatory processes to the development of the early stages of diabetic retinopathy. Exp Diabetes Res 2007:95103. Lee, R, Wang, CC, and Adamis, AP. 1998. Ocular neovascularization: an epidemiologic review. Surv Ophthalmol 43:245-269. Klein, R., Klein, B.E., Moss, S.E., Davis, M.D., and DeMets, D.L. 1984. The Wisconsin epidemiologic study of diabetic retinopathy. II. Prevalence and risk of diabetic retinopathy when age at diagnosis is less than 30 years. Arch Ophthalmol 102:520-526. Klein, R., Klein, B.E., Moss, S.E., Davis, M.D., and DeMets, D.L. 1984. The Wisconsin epidemiologic study of diabetic retinopathy. III. Prevalence and risk of diabetic retinopathy when age at diagnosis is 30 or more years. Arch Ophthalmol 102:527-532. Frank, RN. 2004. Diabetic retinopathy. N Engl J Med 350:48-58. Ishida, S., Usui, T., Yamashiro, K., Kaji, Y., Amano, S., Ogura, Y., Hida, T., Oguchi, Y., Ambati, J., Miller, J.W., et al. 2003. VEGF164-mediated inflammation is required for pathological, but not physiological, ischemia-induced retinal neovascularization. J Exp Med 198:483-489. 44 l3. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. Klein, R., Klein, B.E., Moss, S.E., and Cruickshanks, K]. 1994. The Wisconsin Epidemiologic Study of diabetic retinopathy. XIV. Ten-year incidence and progression of diabetic retinopathy. Arch Ophthalmol 112:1217-1228. Klein, R., Klein, B.E., Moss, S.E., Davis, M.D., and DeMets, D.L. 1984. The Wisconsin epidemiologic study of diabetic retinopathy. IV. Diabetic macular edema. Ophthalmology 91 :1464-1474. Lyons, T.J., Jenkins, A.J., Zheng, D., Lackland, D.T., McGee, D., Garvey, WT, and Klein, R.L. 2004. Diabetic retinopathy and serum lipoprotein subclasses in the DCCT/EDIC cohort. Invest Ophthalmol Vis Sci 45:910-918. Kissebah, A.H., Kohner, E.M., Lewis, B., Siddiq, Y.K., Lowy, C., and Fraser, TR. 1975. Plasma-lipids and glucose/insulin relationship in non-insulin-requiring diabetics with and without retinopathy. Lancet 1:1104-1 108. Ferris, F .L., 3rd, Chew, E.Y., and Hoogwerf, 8.1. 1996. Serum lipids and diabetic retinopathy. Early Treatment Diabetic Retinopathy Study Research Group. Diabetes Care 19:1291-1293. Chew, E.Y., Klein, M.L., Ferris, F.L., 3rd, Remaley, N.A., Murphy, R.P., Chantry, K., Hoogwerf, B.J., and Miller, D. 1996. Association of elevated serum lipid levels with retinal hard exudate in diabetic retinopathy. Early Treatment Diabetic Retinopathy Study (ETDRS) Report 22. Arch Ophthalmol 114:1079- 1084. Van Eck, W.F. 1959. The effect of a low fat diet on the serum lipids in diabetes and its significance in diabetic retinopathy. Am J Med 27:196-21 1. Duncan, L.J., Cullen, J .F ., Ireland, J .T., Nolan, J ., Clarke, B.F., and Oliver, M.F. 1968. A three-year trial of atromid therapy in exudative diabetic retinopathy. Diabetes 17:458-467. Houtsmuller, A.J., Zahn, K.J., and Henkes, HE. 1980. Unsaturated fats and progression of diabetic retinopathy. Doc Ophthalmol 48:363-371. Kreisberg, RA. 1998. Diabetic dyslipidemia. Am J Cardiol 82:67U-73U; discussion 85U-86U. Coppack, S.W., Evans, R.D., Fisher, R.M., Frayn, K.N., Gibbons, G.F., Humphreys, S.M., Kirk, M.L., Potts, J.L., and Hockaday, TD. 1992. Adipose tissue metabolism in obesity: lipase action in vivo before and after a mixed meal. Metabolism 41:264-272. 45 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. Weinstock, P.H., Levak-Frank, S., Hudgins, L.C., Radner, H., Friedman, J.M., Zechner, R., and Breslow, J .L. 1997. Lipoprotein lipase controls fatty acid entry into adipose tissue, but fat mass is preserved by endogenous synthesis in mice deficient in adipose tissue lipoprotein lipase. Proc Natl Acad Sci U S A 94:10261- 10266. Goldberg, RB. 1981. Lipid disorders in diabetes. Diabetes Care 4:561-572. Brenner, RR. 2003. Hormonal modulation of delta6 and delta5 desaturases: case of diabetes. Prostaglandins Leukot Essent Fatty Acids 68: 151-162. Goldberg, U. 2001. Clinical review 124: Diabetic dyslipidemia: causes and consequences. J Clin Endocrinol Metab 86:965-971. Goldberg, RB, and Capuzzi, D. 2001. Lipid disorders in type 1 and type 2 diabetes. Clin Lab Med 21 :147-172, vii. Poisson, J .P. 1989. Essential fatty acid metabolism in diabetes. Nutrition 5:263- 266. Vessby, B. 2000. Dietary fat and insulin action in humans. Br J Nutr 83 Suppl 1:S91-96. Bierman, EL. 1992. George Lyman Duff Memorial Lecture. Atherogenesis in diabetes. Arterioscler T hromb 12:647-656. Syvanne, M., and Taskinen, MR. 1997. Lipids and lipoproteins as coronary risk factors in non-insulin-dependent diabetes mellitus. Lancet 350 Suppl 1:8120-23. Solano, M.P., and Goldberg, RB. 2006. Management of dyslipidemia in diabetes. Cardiol Rev 14:125-135. Stampfer, M.J., Krauss, R.M., Ma, J., Blanche, P.J., Holl, L.G., Sacks, F .M., and Hennekens, CH. 1996. A prospective study of triglyceride level, low-density lipoprotein particle diameter, and risk of myocardial infarction. Jama 276:882- 888. Perez, A., Wagner, A.M., Carreras, G., Gimenez, G., Sanchez-Quesada, J.L., ’ Rigla, M., Gomez-Gerique, J.A., Pou, J .M., and de Leiva, A. 2000. Prevalence and phenotypic distribution of dyslipidemia in type 1 diabetes mellitus: effect of glycemic control. Arch Intern Med 160:2756-2762. Kordonouri, 0., Danne, T., Hopfenmuller, W., Enders, I., Hovener, G., and Weber, B. 1996. Lipid profiles and blood pressure: are they risk factors for the development of early background retinopathy and incipient nephropathy in children with insulin-dependent diabetes mellitus? Acta Paediatr 85:43-48. 46 37. 38. 39. 40. 41. 42. 43. 45. 46. 47. 48. Calder, P.C. 2003. N-3 polyunsaturated fatty acids and inflammation: from molecular biology to the clinic. Lipids 38:343-3 52. Calder, P.C. 2006. n-3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr 83: 1 505S-1519S. Chen, W., Jump, D.B., Esselman, W.J., and Busik, J.V. 2007. Inhibition of cytokine signaling in human retina] endothelial cells through modification of caveolae/lipid rafts by docosahexaenoic acid. Invest Ophthalmol Vis Sci 48:18-26. Chen, W., Esselman, W.J., Jump, DB, and Busik, J .V. 2005. Anti-inflammatory effect of docosahexaenoic acid on cytokine-induced adhesion molecule expression in human retinal vascular endothelial cells. Invest Ophthalmol Vis Sci 46:4342-4347. Simopoulos, AP. 1997. Omega-6/omega-3 fatty acid ratio and trans fatty acids in non-insulin-dependent diabetes mellitus. Ann N Y A cad Sci 827:327-33 8. Simopoulos, AP. 2003. Importance of the ratio of omega-6/omega-3 essential fatty acids: evolutionary aspects. World Rev Nutr Diet 9221-22. Simopoulos, AP. 2008. The importance of the omega-6/omega-3 fatty acid ratio in cardiovascular disease and other chronic diseases. Exp Biol Med (Maywood) 233:674-688. Ferrucci, L., Cherubini, A., Bandinelli, S., Bartali, B., Corsi, A., Lauretani, F., Martin, A., Andres-Lacueva, C., Senin, U., and Guralnik, J .M. 2006. Relationship of plasma polyunsaturated fatty acids to circulating inflammatory markers. J Clin Endocrinol Metab 91:439-446. Harder, T., and Simons, K. 1997. Caveolae, DIGS, and the dynamics of sphingolipid-cholesterol microdomains. Curr Opin Cell Biol 9:534-542. Schroeder, R., London, E., and Brown, D. 1994. Interactions between saturated acyl chains confer detergent resistance on lipids and g1ycosylphosphatidylinositol (GPI)-anchored proteins: GPI-anchored proteins in liposomes and cells Show similar behavior. Proc Natl Acad Sci U S A 91:12130-12134. Holland, W.L., and Summers, SA. 2008. Sphingolipids, insulin resistance, and metabolic disease: new insights from in vivo manipulation of sphingolipid metabolism. Endocr Rev 29:381-402. Summers, SA, and Nelson, DH. 2005. A role for sphingolipids in producing the common features of type 2 diabetes, metabolic syndrome X, and Cushing's syndrome. Diabetes 54: 591 -602. 47 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. Kim, M.Y., Linardic, C., Obeid, L., and Hannun, Y. 1991. Identification of sphingomyelin turnover as an effector mechanism for the action of tumor necrosis factor alpha and gamma-interferon. Specific role in cell differentiation. J Biol Chem 266:484-489. Wiegrnann, K., Schutze, S., Machleidt, T., Witte, D., and Kronke, M. 1994. Functional dichotomy of neutral and acidic Sphingomyelinases in tumor necrosis factor Signaling. Cell 78:1005-1015. Chavez, J.A., Knotts, T.A., Wang, L.P., Li, G., Dobrowsky, R.T., Florant, G.L., and Summers, SA. 2003. A role for ceramide, but not diacylglycerol, in the antagonism of insulin signal transduction by saturated fatty acids. J Biol Chem 278:10297-10303. Chavez, J.A., and Summers, SA. 2003. Characterizing the effects of saturated fatty acids on insulin signaling and ceramide and diacylglycerol accumulation in 3T3-L1 adipocytes and C2C12 myotubes. Arch Biochem Biophys 419:101-109. Turinsky, J., O'Sullivan, D.M., and Bayly, BR 1990. 1,2-Diacylglycerol and ceramide levels in insulin-resistant tissues of the rat in vivo. J Biol Chem 265:16880-16885. Gorska, M., Dobrzyn, A., Zendzian-Piotrowska, M., and Gorski, J. 2004. Effect of streptozotocin-diabetes on the functioning of the Sphingomyelin-signalling pathway in skeletal muscles of the rat. Horm Metab Res 36: 14-21. Adams, J.M., 2nd, Pratipanawatr, T., Berria, R., Wang, E., DeFronzo, R.A., Sullards, M.C., and Mandarino, LJ. 2004. Ceramide content is increased in skeletal muscle from obese insulin-resistant humans. Diabetes 53:25-31. Straczkowski, M., Kowalska, 1., Nikolajuk, A., Dzienis-Straczkowska, S., Kinalska, I., Baranowski, M., Zendzian-Piotrowska, M., Brzezinska, Z., and Gorski, J. 2004. Relationship between insulin sensitivity and sphingomyelin signaling pathway in human skeletal muscle. Diabetes 53: 1215-1221. Straczkowski, M., Kowalska, 1., Baranowski, M., Nikolajuk, A., Otziomek, E., Zabielski, P., Adamska, A., Blachnio, A., Gorski, J., and Gorska, M. 2007. Increased skeletal muscle ceramide level in men at risk of developing type 2 diabetes. Diabetologia 50:2366-23 73. Neschen, S., Moore, 1., Regittnig, W., Yu, C.L., Wang, Y., Pypaert, M., Petersen, K.F., and Shulman, G.I. 2002. Contrasting effects of fish oil and safflower oil on hepatic peroxisomal and tissue lipid content. Am J Physiol Endocrinol Metab 282:E395-401. 48 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. Fox, T.E., Han, X., Kelly, S., Merrill, A.H., 2nd, Martin, R.E., Anderson, R.E., Gardner, T.W., and Kester, M. 2006. Diabetes alters sphingolipid metabolism in the retina: a potential mechanism of cell death in diabetic retinopathy. Diabetes 55:3573-3580. Lecomte, M., Paget, C., Ruggiero, D., Wiemsperger, N., and Lagarde, M. 1996. Docosahexaenoic acid is a major n-3 polyunsaturated fatty acid in bovine retinal microvessels. J Neurochem 66:2 160-2167. Tikhonenko, M., Lydic, T.A., Wang, Y., Chen, W., Opreanu, M., Sochacki, A., McSorley, K.M., Renis, R.L., Kern, T., Jump, D.B., et al. 2009. Remodeling of Retinal Fatty Acids in an Animal Model of Diabetes: a Decrease in Long Chain Polyunsaturated Fatty Acids is Associated with a Decrease in Fatty Acid Elongases Elov12 and Elovl4. Diabetes. SanGiovanni, JP, and Chew, E.Y. 2005. The role of omega-3 long-chain polyunsaturated fatty acids in health and disease of the retina. Prog Retin Eye Res 24287-138. Meyer, A., Kirsch, H., Domergue, F., Abbadi, A., Sperling, P., Bauer, J ., Cirpus, P., Zank, T.K., Moreau, H., Roscoe, T.J., et a1. 2004. Novel fatty acid elongases and their use for the reconstitution of docosahexaenoic acid biosynthesis. J Lipid Res 45: 1 899-1909. Lagali, P.S., Liu, J., Ambasudhan, R., Kakuk, L.E., Bernstein, S.L., Seigel, G.M., Wong, P.W., and Ayyagari, R. 2003. Evolutionarily conserved ELOVL4 gene expression in the vertebrate retina. Invest Ophthalmol Vis Sci 44:2841-2850. Umeda, S., Ayyagari, R., Suzuki, M.T., Ono, F., Iwata, F., Fujiki, K., Kanai, A., Takada, Y., Yoshikawa, Y., Tanaka, Y., et a1. 2003. Molecular cloning of ELOVL4 gene from cynomolgus monkey (Macaca fascicularis). Exp Anim 52:129-135. Zhang, X.M., Yang, Z., Karan, G., Hashimoto, T., Baehr, W., Yang, X.J., and Zhang, K. 2003. Elovl4 mRNA distribution in the developing mouse retina and phylogenetic conservation of Elovl4 genes. Mol Vis 92301-307. Decsi, T., Minda, H., Hermann, R., Kozari, A., Erhardt, E., Burns, 1., Molnar, S., and Soltesz, G. 2002. Polyunsaturated fatty acids in plasma and erythrocyte membrane lipids of diabetic children. Prostaglandins Leukot Essent Fatty Acids 67:203-210. Futterman, S., and Kupfer, C. 1968. The fatty acid composition of the retinal vasculature of normal and diabetic human eyes. Invest Ophthalmol 7:105-108. 49 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. Hartong, D.T., Berson, EL, and Dryja, T.P. 2006. Retinitis pigmentosa. Lancet 368:1795-1809. Hoffinan, DR, and Birch, D.G. 1995. Docosahexaenoic acid in red blood cells of patients with X-linked retinitis pigmentosa. Invest Ophthalmol Vis Sci 3621009- 1018. Ren, H., Magulike, N., Ghebremeskel, K., and Crawford, M. 2006. Primary open- angle glaucoma patients have reduced levels of blood docosahexaenoic and eicosapentaenoic acids. Prostaglandins Leukot Essent Fatty Acids 74: 1 57-163. SanGiovanni, J.P., Chew, E.Y., Clemons, T.E., Davis, M.D., Ferris, F.L., 3rd, Gensler, G.R., Kurinij, N., Lindblad, A.S., Milton, R.C., Seddon, J.M., et al. 2007. The relationship of dietary lipid intake and age-related macular degeneration in a case-control study: AREDS Report No. 20. Arch Ophthalmol 125:671-679. Bazan, N.G. 2006. Cell survival matters: docosahexaenoic acid signaling, neuroprotection and photoreceptors. Trends Neurosci 29:263-271. Connor, K.M., SanGiovanni, J.P., Lofqvist, C., Aderman, C.M., Chen, J., Higuchi, A., Hong, S., Pravda, E.A., Majchrzak, S., Carper, D., et al. 2007. Increased dietary intake of omega-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nat Med 13 :868-873. Powell, ED, and Field, RA. 1964. Diabetic Retinopathy And Rheumatoid Arthritis. Lancet 41:17-18. Joussen, A.M., Poulaki, V., Mitsiades, N., Kirchhof, B., Koizumi, K., Dohmen, S., and Adamis, AP. 2002. Nonsteroidal anti-inflammatory drugs prevent early diabetic retinopathy via TNF-alpha suppression. F aseb J 16:43 8-440. Joussen, A.M., Murata, T., Tsujikawa, A., Kirchhof, B., Bursell, S.E., and Adamis, AP. 2001. Leukocyte-mediated endothelial cell injury and death in the diabetic retina. Am J Pathol 158:147-152. McLeod, D.S., Lefer, D.J., Merges, C., and Lutty, GA. 1995. Enhanced expression of intracellular adhesion molecule-1 and P-selectin in the diabetic human retina and choroid. Am J Pathol 147:642-653. Joussen, A.M., Poulaki, V., Le, M.L., Koizumi, K., Esser, C., Janicki, H., Schraermeyer, U., Kociok, N., Fauser, S., Kirchhof, B., et al. 2004. A central role for inflammation in the pathogenesis of diabetic retinopathy. Faseb J 18:1450- 1452. 50 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. Limb, G.A., Chignell, A.H., Green, W., LeRoy, F., and Dumonde, DC. 1996. Distribution of TNF alpha and its reactive vascular adhesion molecules in fibrovascular membranes of proliferative diabetic retinopathy. Br J Ophthalmol 80:168-173. Murata, T., Nakagawa, K., Khalil, A., Ishibashi, T., lnomata, H., and Sueishi, K. 1996. The relation between expression of vascular endothelial growth factor and breakdown of the blood-retinal barrier in diabetic rat retinas. Lab Invest 74:819- 825. Adamis, A.P., Miller, J .W., Bemal, M.T., D'Amico, D.J., Folkman, J ., Yeo, T.K., and Yeo, K.T. 1994. Increased vascular endothelial growth factor levels in the vitreous of eyes with proliferative diabetic retinopathy. Am J Ophthalmol 118:445-450. Taylor, P.C., Williams, R.O., and Feldmann, M. 2004. Tumour necrosis factor alpha as a therapeutic target for immune-mediated inflammatory diseases. Curr Opin Biotechnol 15:557-563. Eizirik, D.L., and Mandrup-Poulsen, T. 2001. A choice of death--the signal— transduction of immune-mediated beta-cell apoptosis. Diabetologia 44:2115- 2133. Ruan, H., and Lodish, HF. 2003. Insulin resistance in adipose tissue: direct and indirect effects of tumor necrosis factor-alpha. Cytokine Growth Factor Rev 14:447-455. Navarro, J .F., and Mora-Femandez, C. 2006. The role of TNF-alpha in diabetic nephropathy: pathogenic and therapeutic implications. Cytokine Growth Factor Rev 17:441-450. Hawrami, K., Hitrnan, G.A., Rema, M., Snehalatha, C., Viswanathan, M., Rarnachandran, A., and Mohan, V. 1996. An association in non-insulin-dependent diabetes mellitus subjects between susceptibility to retinopathy and tumor necrosis factor polymorphism. Hum Immunol 46:49-54. Limb, G.A., Webster, L., Soomro, H., Janikoun, S., and Shilling, J. 1999. Platelet expression of tumour necrosis factor-alpha (TNF -a1pha), TNF receptors and intercellular adhesion molecule-1 (ICAM-1) in patients with proliferative diabetic retinopathy. Clin Exp Immunol 118:213-218. Spranger, J ., Meyer-Schwickerath, R., Klein, M., Schatz, H., and Pfeiffer, A. 1995. [TNF-alpha level in the vitreous body. Increase in neovascular eye diseases and proliferative diabetic retinopathy]. Med Klin (Munich) 90: 134-137. 51 90. 91. 92. 93. 94. 95. 96. 97. 98. Sfikakis, P.P., Markomichelakis, N., Theodossiadis, G.P., Grigoropoulos, V., Katsilambros, N., and Theodossiadis, PG. 2005. Regression of sight-threatening macular edema in type 2 diabetes following treatment with the anti-tumor necrosis factor monoclonal antibody infliximab. Diabetes Care 28:445-447. Aiello, L.P., Avery, R.L., Arrigg, P.G., Keyt, B.A., Jampel, H.D., Shah, S.T., Pasquale, L.R., Thieme, H., Iwamoto, M.A., Park, J.B., et al. 1994. Vascular endothelial growth factor in ocular fluid of patients with diabetic retinopathy and other retinal disorders. N Engl J Med 331 :1480-1487. Amin, R.H., Frank, R.N., Kennedy, A., Eliott, D., Puklin, J .E., and Abrams, G.W. 1997. Vascular endothelial grth factor is present in glial cells of the retina and optic nerve of human subjects with nonproliferative diabetic retinopathy. Invest Ophthalmol Vis Sci 38:36-47. Miller, J .W., Adamis, A.P., Shima, D.T., D'Amore, P.A., Moulton, R.S., O'Reilly, M.S., Folkman, J., Dvorak, H.F., Brown, L.F., Berse, B., et a1. 1994. Vascular endothelial grth factor/vascular permeability factor is temporally and spatially correlated with ocular angiogenesis in a primate model. Am J Pathol 145:574- 584. Qaum, T., Xu, Q., Joussen, A.M., Clemens, M.W., Qin, W., Miyamoto, K., Hassessian, H., Wiegand, S.J., Rudge, J ., Yancopoulos, G.D., et al. 2001. VEGF- initiated blood-retinal barrier breakdown in early diabetes. Invest Ophthalmol Vis Sci 42:2408-2413. Aiello, L.P., Northrup, J.M., Keyt, B.A., Takagi, H., and Iwamoto, M.A. 1995. Hypoxic regulation of vascular endothelial growth factor in retinal cells. Arch Ophthalmol 11321538-1544. Busik, J.V., Mohr, S., and Grant, MB. 2008. Hyperglycemia-induced reactive oxygen species toxicity to endothelial cells is dependent on paracrine mediators. Diabetes 57:1952-1965. Famiglietti, E.V., Stopa, E.G., McGookin, E.D., Song, P., LeBlanc, V., and Streeten, B.W. 2003. Immunocytochemical localization of vascular endothelial growth factor in neurons and glial cells of human retina. Brain Res 969:195-204. Joussen, A.M., Poulaki, V., Qin, W., Kirchhof, B., Mitsiades, N., Wiegand, S.J., Rudge, J., Yancopoulos, G.D., and Adamis, AP. 2002. Retinal vascular endothelial growth factor induces intercellular adhesion molecule-1 and endothelial nitric oxide synthase expression and initiates early diabetic retinal leukocyte adhesion in vivo. Am J Pathol 160:501-509. 52 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. Senger, D.R., Connolly, D.T., Van de Water, L., Feder, J., and Dvorak, HF. 1990. Purification and NH2-terminal amino acid sequence of guinea pig tumor- secreted vascular permeability factor. Cancer Res 50:1774-1778. Ng, B.W., and Adamis, AP. 2005. Targeting angiogenesis, the underlying disorder in neovascular age-related macular degeneration. Can J Ophthalmol 40:352-368. Gerhardinger, C., Brown, L.F., Roy, S., Mizutani, M., Zucker, C.L., and Lorenzi, M. 1998. Expression of vascular endothelial growth factor in the human retina and in nonproliferative diabetic retinopathy. Am J Pathol 152:1453-1462. Segawa, Y., Shirao, Y., Yamagishi, S., Higashide, T., Kobayashi, M., Katsuno, K., lyobe, A., Harada, H., Sato, F., Miyata, H., et al. 1998. Upregulation of retinal vascular endothelial growth factor mRNAs in Spontaneously diabetic rats without ophthalmoscopic retinopathy. A possible participation of advanced glycation end products in the development of the early phase of diabetic retinopathy. Ophthalmic Res 30:333-339. Aiello, L.P., Pierce, E.A., Foley, E.D., Takagi, H., Chen, H., Riddle, L., Ferrara, N., King, G.L., and Smith, LE. 1995. Suppression of retinal neovascularization in vivo by inhibition of vascular endothelial growth factor (VEGF) using soluble VEGF-receptor chimeric proteins. Proc Natl Acad Sci U S A 92: 10457-10461. Adamis, A.P., Altaweel, M., Bressler, N.M., Cunningham, E.T., Jr., Davis, M.D., Goldbaum, M., Gonzales, C., Guyer, D.R., Barrett, K., and Patel, M. 2006. Changes in retinal neovascularization after pegaptanib (Macugen) therapy in diabetic individuals. Ophthalmology 113:23-28. Arevalo, J .F., Garcia-Amaris, R.A., Roca, J .A., Sanchez, J .G., Wu, L., Berrocal, M.H., and Maia, M. 2007. Primary intravitreal bevacizumab for the management of pseudophakic cystoid macular edema: pilot study of the Pan-American Collaborative Retina Study Group. J Cataract Refract Surg 33:2098-2105. Ng, E.W., and Adamis, AP. 2006. Anti-VEGF aptarner (pegaptanib) therapy for ocular vascular diseases. Ann N Y Acad Sci 1082: 1 51-171. Kowluru, RA, and Odenbach, S. 2004. Role of interleukin-lbeta in the pathogenesis of diabetic retinopathy. Br J Ophthalmol 88: 1343-1347. Vincent, J.A., and Mohr, S. 2007. Inhibition of caspase-l/interleukin-lbeta signaling prevents degeneration of retinal capillaries in diabetes and galactosemia. Diabetes 56:224-230. 53 109. 110. 111. 112. 113. 114. 115. 116. 117. 118. 119. 120. 121. Kowluru, RA, and Odenbach, S. 2004. Role of interleukin-lbeta in the development of retinopathy in rats: effect of antioxidants. Invest Ophthalmol Vis Sci 45:4161-4166. Mohr, S., Xi, X., Tang, J ., and Kern, TS. 2002. Caspase activation in retinas of diabetic and galactosemic mice and diabetic patients. Diabetes 51:1172-1179. Karin, M., and Lin, A. 2002. NF-kappaB at the crossroads of life and death. Nat Immunol 3:221-227. Rothwarf, D.M., and Karin, M. 1999. The NF-kappa B activation pathway: a paradigm in information transfer from membrane to nucleus. Sci ST KE 1999:RE1. Baldwin, A.S., Jr. 2001. Series introduction: the transcription factor NF-kappaB and human disease. J Clin Invest 107:3-6. Kowluru, R.A., Koppolu, P., Chakrabarti, S., and Chen, S. 2003. Diabetes- induced activation of nuclear transcriptional factor in the retina, and its inhibition by antioxidants. Free Radic Res 37:1 169-1180. Romeo, G., Liu, W.H., Asnaghi, V., Kern, TS, and Lorenzi, M. 2002. Activation of nuclear factor-kappaB induced by diabetes and high glucose regulates a proapoptotic program in retinal pericytes. Diabetes 51:2241-2248. Liu, P., and Anderson, R.G. 1995. Compartrnentalized production of ceramide at the cell surface. J Biol Chem 270:27179-27185. Mathias, S., Younes, A., Kan, C.C., Orlow, 1., Joseph, C., and Kolesnick, RN. 1993. Activation of the sphingomyelin Signaling pathway in intact EL4 cells and in a cell-free system by IL-1 beta. Science 259:519-522. Yang, Z., Costanzo, M., Golde, D.W., and Kolesnick, RN. 1993. Tumor necrosis factor activation of the sphingomyelin pathway signals nuclear factor kappa B translocation in intact HL-60 cells. J Biol Chem 268:20520-20523. Marathe, S., Schissel, S.L., Yellin, M.J., Beatini, N., Mintzer, R., Williams, K.J., and Tabas, I. 1998. Human vascular endothelial cells are a rich and regulatable source of secretory sphingomyelinase. Implications for early atherogenesis and ceramide-mediated cell signaling. J Biol Chem 273:4081-4088. Stylianou, E., O'Neill, L.A., Rawlinson, L., Edbrooke, M.R., Woo, P., and Saklatvala, J. 1992. Interleukin 1 induces NF-kappa B through its type I but not its type II receptor in lymphocytes. J Biol Chem 267:15836-15841. Duan, RD. 2006. Alkaline sphingomyelinase: an old enzyme with novel implications. Biochim Biophys Acta 1761:281-291. 54 122. 123. 124. 125. 126. 127. 128. 129. 130. 131. 132. 133. Kolesnick, R. 2002. The therapeutic potential of modulating the ceramide/Sphingomyelin pathway. J Clin Invest 110:3-8. Pavoine, C., and Pecker, F. 2009. Sphingomyelinases: their regulation and roles in cardiovascular pathophysiology. Cardiovasc Res 82: 1 75-1 83. Marchesini, N., and Hannun, Y.A. 2004. Acid and neutral sphingomyelinases: roles and mechanisms of regulation. Biochem Cell Biol 82:27-44. Schneider, P.B., and Kennedy, E.P. 1967. Sphingomyelinase in normal human spleens and in spleens from subjects with Niemann-Pick disease. J Lipid Res 8:202-209. Lin, B., Hassler, D.F., Smith, G.K., Weaver, K., and Hannun, Y.A. 1998. Purification and characterization of a membrane bound neutral pH optimum magnesium-dependent and phosphatidylserine-stimulated sphingomyelinase from rat brain. J Biol Chem 273:34472-34479. Marchesini, N., Luberto, C., and Hannun, Y.A. 2003. Biochemical properties of mammalian neutral Sphingomyelinase 2 and its role in sphingolipid metabolism. J Biol Chem 278:13775-13783. Goni, F.M., and Alonso, A. 2002. Sphingomyelinases: enzymology and membrane activity. F EBS Lett 531:38-46. Levade, T., Malagarie-Cazenave, S., Gouaze, V., Segui, B., Tardy, C., Betito, S., Andrieu-Abadie, N., and Cuvillier, O. 2002. Ceramide in apoptosis: a revisited role. Neurochem Res 27:601-607. Clarke, C.J., and Hannun, Y.A. 2006. Neutral sphingomyelinases and nSMase2: bridging the gaps. Biochim Biophys Acta 1758:1893-1901. Tani, M., and Hannun, Y.A. 2007. Neutral sphingomyelinase 2 is palmitoylated on multiple cysteine residues. Role of palmitoylation in subcellular localization. J Biol Chem 282: 10047-10056. Tani, M., Ito, M., and Igarashi, Y. 2007. Ceramide/sphingosine/sphingosine 1- phosphate metabolism on the cell surface and in the extracellular space. Cell Signal 19:229-237. Veldman, R.J., Maestre, N., Aduib, G.M., Medin, J .A., Salvayre, R., and Levade, T. 2001. A neutral sphingomyelinase resides in sphingolipid-enriched microdomains and is inhibited by the caveolin-scaffolding domain: potential implications in tumour necrosis factor signalling. Biochem J 355:859-868. 55 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. 144. 145. Czamy, M., Liu, J ., Oh, P., and Schnitzer, J .E. 2003. Transient mechanoactivation of neutral sphingomyelinase in caveolae to generate ceramide. J Biol Chem 278:4424-4430. Andrieu-Abadie, N., and Levade, T. 2002. Sphingomyelin hydrolysis during apoptosis. Biochim Biophys Acta 1585:126-134. Pettus, B.J., Chalfant, CE, and Hannun, Y.A. 2002. Ceramide in apoptosis: an overview and current perspectives. Biochim Biophys Acta 1585:] 14-125. Clarke, C.J., Truong, T.G., and Hannun, Y.A. 2007. Role for neutral sphingomyelinase-2 in tumor necrosis factor alpha—stimulated expression of vascular cell adhesion molecule-1 (VCAM) and intercellular adhesion molecule-1 (ICAM) in lung epithelial cells: p38 MAPK is an upstream regulator of nSMase2. J Biol Chem 282:1384-1396. Strum, J .C., Ghosh, S., and Bell, RM. 1997. Lipid second messengers. A role in cell grth regulation and cell cycle progression. Adv Exp Med Biol 407:421-431. Pettus, B.J., Chalfant, CE, and Hannun, Y.A. 2004. Sphingolipids in inflammation: roles and implications. Curr Mol Med 4:405-418. Adam-Klages, S., Adam, D., Wiegmann, K., Struve, S., Kolanus, W., Schneider- Mergener, J., and Kronke, M. 1996. FAN, a novel WD-repeat protein, couples the p55 TNF-receptor to neutral sphingomyelinase. Cell 86:937-947. Segui, B., Andrieu-Abadie, N., Adam-Klages, S., Meilhac, 0., Kreder, D., Garcia, V., Bruno, A.P., Jaffrezou, J.P., Salvayre, R., Kronke, M., et a]. 1999. CD40 signals apoptosis through FAN-regulated activation of the sphingomyelin- ceramide pathway. J Biol Chem 274:37251-3 7258. Segui, B., Cuvillier, O., Adam-Klages, S., Garcia, V., Malagarie-Cazenave, S., Leveque, S., Caspar-Bauguil, S., Coudert, J., Salvayre, R., Kronke, M., et a]. 2001. Involvement of FAN in TNF-induced apoptosis. J Clin Invest 108:143-151. Rethy, LA. 2000. Growth regulation, acid sphingomyelinase gene and genomic imprinting: lessons from an experiment of nature. Pathol Oncol Res 6:298-300. Simonaro, C.M., Park, J.H., Eliyahu, E., Shtraizent, N., McGovem, M.M., and Schuchman, EH. 2006. lrnprinting at the SMPD] locus: implications for acid sphingomyelinase-deficient Niemann-Pick disease. Am J Hum Genet 78:865-870. Ferlinz, K., Hurwitz, R., Moczall, H., Lansmann, S., Schuchman, E.H., and Sandhoff, K. 1997. Functional characterization of the N-glycosylation Sites of human acid sphingomyelinase by site-directed mutagenesis. Eur J Biochem 243:511-517. 56 146. 147. 148. 149. 150. 151. 152. 153. 154. 155. 156. 157. Schuchman, E.H., Levran, 0., Pereira, L.V., and Desnick, R.J. 1992. Structural organization and complete nucleotide sequence of the gene encoding human acid sphingomyelinase (SMPDl). Genomics 12:197-205. Garcia-Barros, M., Paris, F., Cordon-Cardo, C., Lyden, D., Rafii, S., Haimovitz- Friedman, A., Fuks, Z., and Kolesnick, R. 2003. Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science 300:1155-1159. Jensen, J .M., Schutze, S., For], M., Kronke, M., and Proksch, E. 1999. Roles for tumor necrosis factor receptor p55 and Sphingomyelinase in repairing the cutaneous permeability barrier. J Clin Invest 104: 1761-1770. Chavakis, E., and Dimmeler, S. 2002. Regulation of endothelial cell survival and apoptosis during angiogenesis. Arterioscler T hromb Vasc Biol 22:887-893. Grassme, H., Schwarz, H., and Gulbins, E. 2001. Molecular mechanisms of ceramide-mediated CD95 clustering. Biochem Biophys Res Commun 284:1016- 1030. Schissel, S.L., Keesler, G.A., Schuchman, E.H., Williams, K.J., and Tabas, I. 1998. The cellular trafficking and zinc dependence of secretory and lysosomal sphingomyelinase, two products of the acid sphingomyelinase gene. J Biol Chem 273:18250-18259. Schissel, S.L., Schuchman, E.H., Williams, K.J., and Tabas, 1. 1996. Zn2+- stimulated sphingomyelinase is secreted by many cell types and is a product of the acid sphingomyelinase gene. J Biol Chem 271:18431-18436. Dobrowsky, RT. 2000. Sphingolipid signalling domains floating on rafts or buried in caves? Cell Signal 12:81-90. Gulbins, E. 2003. Regulation of death receptor signaling and apoptosis by ceramide. Pharmacol Res 47:393-399. Grassme, H., Riethmuller, J., and Gulbins,. E. 2007. Biological aspects of ceramide-enriched membrane domains. Prog Lipid Res 46:161-170. Grassme, H., Jekle, A., Riehle, A., Schwarz, H., Berger, J., Sandhoff, K., Kolesnick, R., and Gulbins, E. 2001. CD95 Signaling via ceramide-rich membrane rafts. J Biol Chem 276:20589-20596. Zeidan, Y.H., and Hannun, Y.A. 2007. Activation of acid sphingomyelinase by protein kinase Cdelta-mediated phosphorylation. J Biol Chem 282:] 1549-11561. 57 158. 159. 160. 161. 162. 163. 164. 165. 166. 167. 168. 169. Callahan, J.W., Jones, C.S., Davidson, D.J., and Shankaran, P. 1983. The active site of lysosomal sphingomyelinase: evidence for the involvement of hydrophobic and ionic groups. J Neurosci Res 10:151-163. Schissel, S.L., Jiang, X., Tweedie-Hardman, J ., Jeong, T., Camejo, E.H., Najib, J ., Rapp, J.H., Williams, K.J., and Tabas, I. 1998. Secretory sphingomyelinase, a product of the acid sphingomyelinase gene, can hydrolyze atherogenic lipoproteins at neutral pH. Implications for atherosclerotic lesion development. J Biol Chem 273:2738-2746. Tabas, I. 1999. Secretory sphingomyelinase. Chem Phys Lipids 102:123-130. Schutze, S., Machleidt, T., and Kronke, M. 1992. Mechanisms of tumor necrosis factor action. Semin Oncol 19:16-24. Schutze, S., Machleidt, T., and Kronke, M. 1994. The role of diacylglycerol and ceramide in tumor necrosis factor and interleukin-1 signal transduction. J Leukoc Biol 56:533-541. Cifone, M.G., Roncaioli, P., De Maria, R., Camarda, G., Santoni, A., Ruberti, G., and Testi, R. 1995. Multiple pathways originate at the FaS/APO-l (CD95) receptor: sequential involvement of phosphatidylcholine-specific phospholipase C and acidic sphingomyelinase in the propagation of the apoptotic signal. Embo J 14:5859-5868. Gomez-Munoz, A., Kong, J ., Salh, B., and Steinbrecher, U.P. 2003. Sphingosine- l-phosphate inhibits acid sphingomyelinase and blocks apoptosis in macrophages. FEBS Lett 539:56-60. Schwandner, R., Wiegrnann, K., Bernardo, K., Kreder, D., and Kronke, M. 1998. TNF receptor death domain-associated proteins T RADD and FADD signal activation of acid sphingomyelinase. J Biol Chem 273:5916-5922. Bilderback, T.R., Gazula, V.R., and Dobrowsky, RT. 2001. Phosphoinositide 3- kinase regulates crosstalk between Trk A tyrosine kinase and p75(NTR)- dependent sphingolipid signaling pathways. J Neurochem 76: 1 540-1551. Dbaibo, G.S., Pushkareva, M.Y., Rachid, R.A., Alter, N., Smyth, M.J., Obeid, L.M., and Hannun, Y.A. 1998. p53-dependent ceramide response to genotoxic stress. J Clin Invest 1022329-339. Quintem, LE, and Sandhoff, K. 1991. Human acid sphingomyelinase from human urine. Methods Enzymol 197:536-540. De Nadai, C., Sestili, P., Cantoni, 0., Lievremont, J.P., Sciorati, C., Barsacchi, R., Moncada, S., Meldolesi, J., and Clementi, E. 2000. Nitric oxide inhibits tumor 58 170. 171. 172. 173. 174. 175. 176. 177. 178. 179. 180. necrosis factor-alpha-induced apoptosis by reducing the generation of ceramide. Proc Natl Acad Sci U S A 97:5480-5485. Santana, P., Pena, L.A., Haimovitz-Friedman, A., Martin, S., Green, D., McLoughlin, M., Cordon-Cardo, C., Schuchman, E.H., Fuks, Z., and Kolesnick, R. 1996. Acid sphingomyelinase-deficient human lyrnphoblasts and mice are defective in radiation-induced apoptosis. Cell 86: 1 89-199. Smith, E.L., and Schuchman, EH. 2008. Acid sphingomyelinase overexpression enhances the antineoplastic effects of irradiation in vitro and in vivo. Mol T her 16:1565-1571. Grammatikos, G., Teichgraber, V., Carpinteiro, A., Trarbach, T., Weller, M., Hengge, U.R., and Gulbins, E. 2007. Overexpression of acid sphingomyelinase sensitizes glioma cells to chemotherapy. Antioxid Redox Signal 9: 1449-1456. Llacuna, L., Mari, M., Garcia-Ruiz, C., Fernandez-Checa, J .C., and Morales, A. 2006. Critical role of acidic sphingomyelinase in murine hepatic ischemia- reperfusion injury. Hepatology 44:561-572. Zumbansen, M., and Stoffe], W. 1997 . Tumor necrosis factor alpha activates NF- kappaB in acid sphingomyelinase-deficient mouse embryonic fibroblasts. J Biol Chem 272:10904-10909. Lin, T., Genestier, L., Pinkoski, M.J., Castro, A., Nicholas, 8., Mogil, R., Paris, F., Fuks, Z., Schuchman, E.H., Kolesnick, R.N., et a]. 2000. Role of acidic Sphingomyelinase in Fas/CD95-mediated cell death. J Biol Chem 275:8657-8663. Schuchman, EH. 2009. The pathogenesis and treatment of acid sphingomyelinase-deficient Niemann-Pick disease. Int J Clin Pharmacol Ther 47 Suppl 1:S48-57. Schuchman, EH. 2009. Acid sphingomyelinase, cell membranes and human disease: Lessons from Niemann-Pick disease. FEBS Lett. Grassme, H., Jendrossek, V., Bock, J., Riehle, A., and Gulbins, E. 2002. Ceramide-rich membrane rafts mediate CD40 clustering. J Immunol 168:298-307. Gulbins, E., and Kolesnick, R. 2003. Raft ceramide in molecular medicine. Oncogene 22:7070-7077. Gulbins, E., and Li, PL. 2006. Physiological and pathophysiological aspects of ceramide. Am J Physiol Regul Integr Comp Physiol 290:R1 1-26. 59 181. 182. 183. 184. 185. 186. 187. 188. 189. 190. 191. 192. Brown, DA, and London, E. 1997. Structure of detergent-resistant membrane domains: does phase separation occur in biological membranes? Biochem Biophys Res Commun 240:1-7. Hooper, NM. 1998. Membrane biology: do glycolipid microdomains really exist? Curr Biol 82R114-116. Schroeder, R.J., Ahmed, S.N., Zhu, Y., London, E., and Brown, DA. 1998. Cholesterol and sphingolipid enhance the Triton X-100 insolubility of glycosylphosphatidylinositol-anchored proteins by promoting the formation of detergent-insoluble ordered membrane domains. J Biol Chem 273:] 150-1157. Palade, GE. 1953. Fine structure of blood capillaries. J Appl Physiol 2421424— 1436. Schnitzer, J.E., McIntosh, D.P., Dvorak, A.M., Liu, J., and Oh, P. 1995. Separation of caveolae from associated microdomains of GPI-anchored proteins. Science 269: 1435-1439. Frank, P.G., Woodman, S.E., Park, D.S., and Lisanti, M.F. 2003. Caveolin, caveolae, and endothelial cell function. Arterioscler Thromb Vasc Biol 23:1161- 1168. Miyawaki-Shimizu, K., Predescu, D., Shimizu, J ., Broman, M., Predescu, S., and Malik, AB. 2006. siRNA-induced caveolin-1 knockdown in mice increases lung vascular permeability via the junctional pathway. Am J Physiol Lung Cell Mol Physiol 290:L405-413. Li, 8., Song, KS, and Lisanti, MP. 1996. Expression and characterization of recombinant caveolin. Purification by polyhistidine tagging and cholesterol- dependent incorporation into defined lipid membranes. J Biol Chem 271:568-573. Murata, M., Peranen, J., Schreiner, R., Wieland, F., Kurzchalia, T.V., and Simons, K. 1995. VIP21/caveolin is a cholesterol-binding protein. Proc Natl Acad Sci U S A 92:10339-10343. Anderson, R.G. 1993. Caveolae: where incoming and outgoing messengers meet. Proc Natl Acad Sci U S A 90:10909-10913. Lisanti, M.P., Tang, Z., Scherer, P.E., Kubler, E., Koleske, A.J., and Sargiacomo, M. 1995. Caveolae, transmembrane signalling and cellular transformation. M01 Membr Biol 12:121-124. Anderson, R.G., and Jacobson, K. 2002. A role for lipid shells in targeting proteins to caveolae, rafts, and other lipid domains. Science 296:1821-1825. 6O 193. 194. 195. 196. 197. 198. 199. 200. 201. 202. 203. 204. 205. Razani, B., and Lisanti, MP. 2001. Caveolins and caveolae: molecular and fimctional relationships. Exp Cell Res 271236-44. Edidin, M. 2003. The state of lipid rafts: from model membranes to cells. Annu Rev Biophys Biomol Struct 32:257-283. Sargiacomo, M., Sudol, M., Tang, Z., and Lisanti, M.F. 1993. Signal transducing molecules and g1ycosyl-phosphatidylinositol-linked proteins form a caveolin-rich insoluble complex in MDCK cells. J Cell Biol 122:789-807. Smart, B.J., Graf, G.A., McNiven, M.A., Sessa, W.C., Engelman, J.A., Scherer, P.B., Okarnoto, T., and Lisanti, M.F. 1999. Caveolins, liquid-ordered domains, and signal transduction. Mol Cell Biol 19:7289-7304. Smart, E.J., Ying, Y.S., Mineo, C., and Anderson, R.G. 1995. A detergent-free method for purifying caveolae membrane from tissue culture cells. Proc Natl Acad Sci USA 92:10104-10108. Fiedler, K., Parton, R.G., Kellner, R., Etzold, T., and Simons, K. 1994. VIP36, a novel component of glycolipid rafts and exocytic carrier vesicles in epithelial cells. EmboJ 13:1729-1740. Harman, L.A., Lisanti, M.P., Rodriguez-Boulan, E., and Edidin, M. 1993. Correctly sorted molecules of a GPI-anchored protein are clustered and immobile when they arrive at the apical surface of MDCK cells. J Cell Biol 120:353-358. Zhang, F ., Crise, B., Su, 3., Hon, Y., Rose, J .K., Bothwell, A., and Jacobson, K. 1991. Lateral diffusion of membrane-spanning and g1ycosylphoSphatidylinositol- linked proteins: toward establishing rules governing the lateral mobility of membrane proteins. J Cell Biol 115:75-84. Kolesnick, R.N., Goni, F.M., and Alonso, A. 2000. Compartrnentalization of ceramide signaling: physical foundations and biological effects. J Cell Physiol 184:285-300. Simons, K., and Ikonen, E. 1997. Functional rafts in cell membranes. Nature 387:569-572. Mathias, S., Pena, LA, and Kolesnick, RN. 1998. Signal transduction of stress via ceramide. Biochem J 335 ( Pt 3):465-480. Ballou, L.R., Laulederkind, S.J., Rosloniec, B.F., and Raghow, R. 1996. Ceramide signalling and the immune response. Biochim Biophys Acta 1301:273-287. Hannun, Y.A. 1996. Functions of ceramide in coordinating cellular responses to stress. Science 274: 1 855-1 859. 61 206. 207. 208. 209. 210. 211. 212. 213. 214. 215. 216. 217. Pena, L.A., Fuks, Z., and Kolesnick, R. 1997. Stress-induced apoptosis and the sphingomyelin pathway. Biochem Pharmacol 53 :61 5-621. Bilderback, T.R., Gazula, V.R., Lisanti, M.P., and Dobrowsky, RT. 1999. Caveolin interacts with Trk A and p75(NTR) and regulates neurotrophin signaling pathways. J Biol Chem 274:257-263. Kolesnick, R., and Hannun, Y.A. 1999. Ceramide and apoptosis. Trends Biochem Sci 24:224-225; author reply 227. Stancevic, B., and Kolesnick, R. 2010. Ceramide-rich platforms in transmembrane Signaling. F EBS Lett. Holopainen, J .M., Subrarnanian, M., and Kinnunen, PK. 1998. Sphingomyelinase induces lipid microdomain formation in a fluid phosphatidylcholine/sphingomyelin membrane. Biochemistry 37: 1 7562-17570. Veiga, M.P., Arrondo, J.L., Goni, F.M., and Alonso, A. 1999. Cerarnides in phospholipid membranes: effects on bilayer stability and transition to nonlamellar phases. Biophys J 76:342-350. Nurminen, T.A., Holopainen, J.M., Zhao, H., and Kinnunen, PK. 2002. Observation of topical catalysis by sphingomyelinase coupled to microspheres. J Am Chem Soc 124:12129-12134. Abdel Shakor, A.B., Kwiatkowska, K., and Sobota, A. 2004. Cell surface ceramide generation precedes and controls F cgammaRII clustering and phosphorylation in rafts. J Biol Chem 279:36778-36787. Dumitru, CA, and Gulbins, E. 2006. TRAIL activates acid sphingomyelinase via a redox mechanism and releases ceramide to trigger apoptosis. Oncogene 25:5612-5625. Grassme, H., Bock, J ., Kun, J ., and Gulbins, E. 2002. Clustering of CD40 ligand is required to form a functional contact with CD40. J Biol Chem 277:30289- 30299. Natoli, G., Costanzo, A., Guido, F., Moretti, F., and Levrero, M. 1998. Apoptotic, non-apoptotic, and anti-apoptotic pathways of tumor necrosis factor signalling. Biochem Pharmacol 56:915-920. Schutze, S., Potthoff, K., Machleidt, T., Berkovic, D., Wiegrnann, K., and Kronke, M. 1992. TNF activates NF-kappa B by phosphatidylcholine-specific phospholipase C-induced "acidic" sphingomyelin breakdown. Cell 71:765-776. 62 218. 219. 220. 221. 222. 223. 224. 225. 226. 227. 228. Rosenman, S.J., Ganji, A.A., Tedder, T.P., and Gallatin, W.M. 1993. Syn-capping of human T lymphocyte adhesion/activation molecules and their redistribution during interaction with endothelial cells. J Leukoc Biol 53:1-10. Simarro, M., Calvo, J ., Vila, J .M., Places, L., Padilla, O., Alberola-Ila, J ., Vives, J ., and Lozano, F. 1999. Signaling through CD5 involves acidic Sphingomyelinase, protein kinase C-zeta, mitogen-activated protein kinase kinase, and c-Jun NHZ-termina] kinase. J Immunol 162:5149-5155. Bezombes, C., Grazide, S., Garret, C., Fabre, C., Quillet-Mary, A., Muller, S., Jaffrezou, J .P., and Laurent, G. 2004. Rituximab antiproliferative effect in B- lymphoma cells is associated with acid-Sphingomyelinase activation in raft microdomains. Blood 104:] 166-1 173. K0, Y.G., Lee, J.S., Kang, Y.S., Ahn, J.H., and Seo, J.S. 1999. TNF-alpha- mediated apoptosis is initiated in caveolae-like domains. J Immunol 162:7217- 7223. Martin, M.U., and Wesche, H. 2002. Summary and comparison of the signaling mechanisms of the Toll/interleukin-l receptor family. Biochim Biophys Acta 1592:265-280. Delton-Vandenbroucke, I., Grammas, P., and Anderson, RE. 1997. Polyunsaturated fatty acid metabolism in retina] and cerebral microvascular endothelial cells. J Lipid Res 38:147-159. Lisanti, M.P., Scherer, P.B., Vidugiriene, J ., Tang, Z., Hermanowski-Vosatka, A., Tu, Y.H., Cook, RR, and Sargiacomo, M. 1994. Characterization of caveolin- rich membrane domains isolated fi'om an endothelial-rich source: implications for human disease. J Cell Biol 126:1] 1-126. Ma, D.W., Seo, J., Davidson, L.A., Callaway, E.S., Fan, Y.Y., Lupton, J.R., and Chapkin, RS. 2004. n-3 PUFA alter caveolae lipid composition and resident protein localization in mouse colon. Faseb J 18: 1 040-1042. Stoffel, W., Jenke, B., Block, B., Zumbansen, M., and Koebke, J. 2005. Neutral Sphingomyelinase 2 (smpd3) in the control of postnatal grth and development. Proc Natl Acad Sci U S A 10214554-4559. Yang, Y., Yin, J ., Baumgartner, W., Samapati, R., Solymosi, E.A., Reppien, E., Kuebler, W.M., and Uhlig, S. 2009. Platelet-activating factor reduces endothelial NO production - role of acid sphingomyelinase. Eur Respir J. Jin, S., Zhang, Y., Yi, F., and Li, PL. 2008. Critical role of lipid raft redox signaling platforms in endostatin-induced coronary endothelial dysfunction. Arterioscler T hromb Vasc Biol 28:485-490. 63 229. 230. 231. 232. 233. Bao, J.X., Xia, M., Poklis, J.L., Han, W.Q., Brimson, C., and Li, PL. 2010. Triggering role of acid sphingomyelinase in endothelial lysosome-membrane firsion and dysfunction in coronary arteries. Am J Physiol Heart Circ Physiol 298:H992-H1002. Zhang, A.Y., Yi, F., Jin, S., Xia, M., Chen, Q.Z., Gulbins, E., and Li, PL. 2007. Acid sphingomyelinase and its redox amplification in formation of lipid raft redox signaling platforms in endothelial cells. Antioxid Redox Signal 9:817-828. Gorska, M., Baranczuk, E., and Dobrzyn, A. 2003. Secretory Zn2+-dependent sphingomyelinase activity in the serum of patients with type 2 diabetes is elevated. Harm Metab Res 35:506-507. Samad, F., Hester, K.D., Yang, Q, Hannun, Y.A., and Bielawski, J. 2006. Altered adipose and plasma sphingolipid metabolism in obesity: a potential mechanism for cardiovascular and metabolic risk. Diabetes 55:2579-2587. Summers, SA. 2006. Ceramides in insulin resistance and lipotoxicity. Prog Lipid Res 45:42-72. 64 Chapter II Docosahexaenoic Acid Inhibits Cytokine Signaling by Downregulating Sphingomyelinases in Human Retinal Endothelial Cells 1. Abstract The anti-inflammatory mechanism of docosahexaenoic acid (DHA) is well accepted; however, the molecular mechanism(s) of DHA action are still not well defined. We have previously demonstrated that DHA modifies the caveolar membrane microdomain and inhibits cytokine-induced inflammation in human retina] endothelial cells (HREC), the resident vasculature affected by diabetic retinopathy. Sphingolipids represent a major component of plasma membrane microdomains and ceramide-enriched microdomains appear to be a prerequisite for inflammatory cytokines signaling. Acid and neutral sphingomyelinase (ASMase and NSMase) are key regulatory enzymes of sphingolipid metabolism, promoting sphingomyelin hydrolysis to pro-inflammatory ceramide. In this study, we propose to test the hypothesis that DHA inhibits cytokine- induced inflammatory Signaling in HREC by downregulating sphingomyelinases. Primary cultures of HREC were maintained in the presence or absence of DHA. ASMase and NSMase activity was determined by sphingomyelinase assay. The expression of ASMase, NSMase, ICAM-1 and VCAM-l was assessed by quantitative PCR and Western blotting. Gene silencing of ASMase and NSMase was obtained by siRNA treatment. Inflammatory cytokines TNFa and IL-IB induced cellular adhesion molecules (CAMS) expression and rapid increase in ASMase and NSMase activity in 65 HREC. Growth of HREC in media supplemented with DHA decreased basal and cytokine-induced ASMase and NSMase expression and activity, as well as upregulation of CAMS expression. Anti-inflammatory effects of DHA on cytokine-induced CAMS expression were mimicked by inhibition/gene silencing of ASMase and NSMase. Inhibition of sphingomyelinases activity was mediated by DHA-induced cholesterol displacement from caveolae/lipid microdomains. Sphingomyelinases pathway rather than ceramide de nova synthesis pathway was important for inflammatory Si gnaling in HREC. This study provides a novel potential mechanism for anti-inflammatory effect of DHA in HREC. DHA downregulates basal and cytokine-induced ASMase and NSMase activity and expression level in HREC and inhibition of sphingomyelinases in endothelial cells prevents cytokine-induced inflammatory response. 66 2. Introduction The retina has a unique fatty acid profile, with the highest level of polyunsaturated fatty acids (PUFAS) in the body, especially DHA (1). Deficiency in DHA has been documented to be associated with a number of retinal degenerative diseases, including retinitis pigmentosa, retinopathy of prematurity and age-related macular degeneration (reviewed in (2)). DHA is also Significantly decreased in plasma of diabetic children (3), as well as in the human retina of diabetic eyes (4). Recent data showed a beneficial effect of dietary DHA in reducing pathological retinal angiogenesis and thus preventing the development of oxygen-induced retinopathy (5). Yet, the DHA protective mechanism in retinopathy remains poorly understood. The aim of our study was to investigate the anti-inflammatory mechanism of DHA in human retina] endothelial cells (HREC), the target tissue affected by diabetic retinopathy. Very early stage diabetic retinopathy is hypothesized to represent a low- grade chronic inflammatory disease that involves leukocyte adhesion to the retinal vasculature, a process mediated by adhesion molecules expressed on the endothelial cell surface, especially intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-l) (6-8). Pro-inflammatory cytokines, including TNFa and IL-lB are increased in diabetic eyes (9-11) and induce upregulation of adhesion molecule expression (12). Our group has previously shown that DHA inhibits cytokine-induced cellular adhesion molecule expression in HREC through cholesterol displacement from caveolae/lipid rafts membrane microdomains (12). HREC contain lipid rafts and Specific plasma membrane microdomains known as caveolae that are considered to have an important role in regulating vascular permeability 67 (13), lipid trafficking, cholesterol homeostasis (14-15) and signal transduction (16-17). The caveolae are stabilized by membrane spanning proteins, the caveolins, a family of palmitoylated hairpin-like membrane proteins (18-21). Caveolin-1, the primary structural protein constituent of caveolae in endothelial cells, is a biochemical marker for caveolae identification (22). Caveolae/lipid rafts are compositionally and functionally specific domains which are not static but are rather in equilibrium with the rest of membrane lipids; they are lateral assemblies of cholesterol, sphingolipids (sphingomyelin, ceramide) and glycerophospholipids (23). Sphingolipids play essential roles in membrane microdomain structure and are now known to also act as messengers in signaling pathways mediating inflammation, apoptosis, cell differentiation and proliferation (24). Ceramides can be generated at the membrane level by catabolism of sphingomyelin by either neutral or acid sphingomyelinases or by de nova synthesis in the endoplasmic reticulum. NSMase and ASMase are rapidly activated by diverse stress stimuli and promote hydrolysis of sphingomyelin into ceramide and phosphorylcholine (25-27). NSMase is considered one of the major candidates for mediating stress-induced production of ceramide; it is a membrane-bound protein and palmitoylated on five cysteine residues via thioester bonds (28-30). ASMase was initially described as a lysosomal enzyme because of its optimal pH at 4.5—5.0. Nevertheless, an ASMase isofonn was recently Shown to be enclosed into secretory vesicles in close proximity to the plasma membrane and to be secreted into the extracellular Space upon cell stimulation (31-33). Liu and Anderson described the plasma membrane form of ASMase in caveolae and Showed that the pro- inflammatory cytokine IL-lB may induce ASMase activation in this compartment (27). 68 Lipid analysis demonstrated that approximately 70% of total cellular sphingomyelin is found in membrane microdomains (20, 34-37). Enrichment of sphingomyelin content in membrane microdomains makes them potential substrate pools for cellular sphingomyelinases to produce a high local concentration of ceramide; ceramide-enriched membrane microdomains have the ability to self-associate to form ceramide rich macrodomains (38-41) that mediate receptor clustering and downstream signaling (42-43). Clustering is shown to be an important feature used by several receptors that mediate inflammatory signaling pathways in diabetic retinopathy, such as the TNFa and IL-IB pathways (44-45). In this study, we tested whether the anti-inflammatory mechanism of DHA is mediated through downregulation of ASMase and NSMase, as manifested by decreased cytokine signaling in human retina] endothelial cells. 69 3. Materials and methods 3.1. Reagents and Supplies DMEM and F12 culture medium, antibiotics, fetal bovine serum and trypsin were obtained from Invitrogen (Carlsbad, CA). Amplex Red Sphingomyelinase Assay Kit, Amplex Red Cholesterol Assay Kit, NuPAGE Novex 10% Bis- Tris gels, Platinum SYBR Green qPCR SuperMix-UDG w/ROX were purchased from Invitrogen (Carlsbad, CA). GW4869 was purchased fiom Calbiochem (San Diego, CA). Desiprarnine, methyl- cyclodextrine and commonly used chemicals and reagents were purchased from Sigma (St. Louis, MO). Caveolin-1 and LAMP] antibodies (BD Bioscience, San Jose, CA), ICAM-l, VCAM-l and NSMase antibodies (Santa Cruz Biotechnology, Santa Cruz, CA) were used. ASMase antibody was a generous gift from Dr. Richard Kolesnick. TNFa and IL-IB were from R&Dsystems (Minneapolis, MN). ON-TARGET plus SMART poo] SMPDl (ASMase), ON-TARGET plus SMART pool SMPD2 (NSMase) and ON- TARGET plus siCONTROL (non-targeting pool, against luciferase) were purchased fi'om Dharmacon (Chicago, IL). For lipid extraction and mass spectrometry, all solvents used were HPLC grade. Methanol (MeOH), and water were purchased from IT Baker (Phillipsburg, NJ). Ammonium hydroxide (NH4OH) and chloroform (CHC13) were from EMD Chemicals (Gibbstown, NJ). Isopropanol was from Fisher Scientific (Pittsburgh, PA). Lipid standards were obtained fiom Avanti Polar Lipids (Alabaster, AL). 70 3.2. Cell Culture Primary cultures of HREC were prepared from tissue (National Disease Research Interchange, Philadelphia, PA) cultured as previously described (46). Passages 1 to 5 were used in the experiments. For experimental treatment, cells were transferred to serum-free medium for 14 to 24 hours before stimulatory agents were added. 3.3. Fatty Acid Treatment Fatty acid stocks were prepared by dissolving fatty acids (NuCheck Prep, Inc., Elysian, MN) in ethanol to a final concentration of 100 mM fatty acid, as described previously (12, 47). The fatty acid stock solutions were diluted to 50 to 100 uM in serum-free medium containing 10 to 20 uM charcoal-treated, solvent-extracted, fatty acid—free bovine serum albumin (BSA; Serologica Inc., Norcross, GA) as a fatty acid carrier. The fatty acid/albumin molar ratio was maintained at 5:1 (48). The final concentration of ethanol in the media was less than 0.1%. Cells were incubated for the times indicated in Results. Equivalent amounts of BSA and ethanol were added to control plates. Linoleic acid, a (06 polyunsaturated fatty acid, was used as a lipid control for the following considerations. The most abundant polyunsaturated fatty acid classes in the retina microvessels are 033 (especially DHA) and (b6 (linoleic acid, arachidonic acid) fatty acids (1). In this study we wanted to compare the effects of the principal 033 PUFA (DHA) with a (06 PUFA (linoleic acid). 71 3.4. Sphingomyelinase Assay Fatty acids (DHA and linoleic acid) were added to HREC in serum-free medium to a concentration of 100 uM fatty acid with 20 uM charcoal-treated, solvent extracted, fatty- acid-free bovine serum albumin (BSA) as a fatty acid carrier. Cells were incubated with fatty acids for specified times at 37°C; equivalent amounts of BSA and ethanol were added to the control plates. Then cells were lysed in the acid lysis buffer (50 mM Sodium Acetate, pH=5; 1% TritonX-l 00; 1 mM EDTA) or neutral lysis buffer (20 mM Tris-HCl, pH=7.5; 1% TritonX-IOO; 1 mM EDTA) with fieshly added protease inhibitor cocktail (Sigma, St. Louis, MO). For the sphingomyelinase assay in a cell-fiee system, 0.5mU of bacterial sphingomyelinase was incubated with 5-80 uM fatty acids (DHA and linoleic acid), while equivalent amounts of ethanol were added to the control wells. Sphingomyelinase activity was measured using the Amplex Red Sphingomyelinase Assay Kit (Molecular Probes, Eugene, OR) as described in the manufacturer’s protocol. 3.5. Methyl-Il-cyclodextrin (MCD) and Cholesterol Treatment To deplete cholesterol from the membranes, cells were treated with 8 mM MCD for 30 minutes. To replenish cholesterol, cells were treated with 25 uM water-soluble cholesterol (25 uM cholesterol complexed with 250 uM MCD; Sigma) for 30 minutes. After MCD or water-soluble cholesterol treatment, the cells were washed twice with PBS containing 40 uM BSA before membrane microdomain isolation, and cell lysate and plasma membrane preparation for the sphingomyelinase assay. 72 3.6. Cholesterol Measurement Cholesterol levels of membrane microdomains isolated from HREC with or without MCD treatment (previously described) were measured by the Amplex Red Cholesterol Assay Kit (Molecular Probes, Eugene, OR) as described in the manufacturer’s protocol. 3.7. Western Blot Analysis Cells were lysed in the lysis buffer (50 mM HEPES, pH 7.5, 150 mM NaCl, 1.5 mM MgC12, 1 mM EGTA, 1% Triton X-100 and 10% glycerol) with fieshly added protease inhibitor cocktail (Sigma) and phosphatase inhibitors (1 mM Na3VO4, 100 uM glycerophosphate, 10 mM NaF, lmM Na4PPi). Protein concentration was measured by Qubit fluorometer (Invitrogen, Eugene, OR) as described in the manufacturer protocol. Proteins were resolved by NuPAGE on Novex 10% Bis-Tris gels, transferred to nitrocellulose membrane, and then immunobloted using appropriate primary antibodies followed by secondary horseradish peroxidase-conjugated antibody (Bio-Rad, Hercules, CA) or lRDye infrared secondary antibodies (Invitrogen, Molecular probes, Eugene, OR). Irnmunoreactive bands were visualized by enhanced chemiluminescence (ECL kit; Amersham Pharrnacia Biotech, Piscataway, NJ) or the Odyssey program. Blots were quantified by scanning densitometry (lmageJ software). 73 3.8. Gene Silencing of ASMase and NSMase For silencing ASMase and NSMase expression, cultured HREC were detached with trypsin, centrifuged at 100 g for 5 minutes and resuspended in electroporation solution (Amaxa, Gaithersburg, MD) to a final concentration of 4-5x105 cells/ 100 pL. The 100 uL of cell suspension was mixed with 100 nM ASMase/NSMase siRNA into the electroporation cuvette. HREC were then electroporated with the Nucleofactor program M-030 (Amaxa Biosystems). The electroporated cells were maintained in supplemented medium in 37°C/5% C02 incubator for 48 hours prior to TNFa (10 ng/ml) and IL-IB (5 ng/ml) treatment. To determine the efficiency of gene silencing, RNA was extracted from 100 nM siRNA treated HREC and used as template for real time-PCR as described (49). ASMase/ NSMase siRNA treatment of HREC induced gene silencing of 94 % for ASMase and 86 % for NSMase. 3.9. Quantitative Real Time-Polymerase Chain Reaction (qRT-PCR) Total RNA was extracted from HREC with or without DHA pretreatment(50). Specific primers for each gene were designed using IDT DNA PrimerQuest software (Coralville, IA): for NSMase: forward —GAGCGCAATCATGGCAGT and reverse —CCCACTGA ACCAGTCACCAT; for ASMase: forward —CAACCTCGCGCTGAAGAA and reverse-— TCCACCATGTCATCCTCAAA. First strand cDNA was synthesized using the SuperScript II RNase H-Reverse Transcriptase (Invitrogen Carlsbad, CA). Synthesized cDNA was mixed with 2x SYBR Green PCR Master Mix (Invitrogen Carlsbad, CA) and different sets of gene-specific forward and reverse primers, and then subjected to real- 74 time PCR quantification using the AB] PRISM 7900 Sequence Detection System (Applied Biosystems). All reactions were performed in triplicates. The relative amounts of mRNAs were calculated by using the comparative CT method (User Bulletin #2, Applied Biosystems). Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRNA. 3.10. Isolation of Caveolae/Lipid Rafts Membrane Domains Isolation of caveolae/lipid raft-enriched detergent-resistant membrane domains were prepared using a slightly modified sucrose gradient ultracentrifugation protocol(47). Briefly, 5 X 106 HREC were washed twice with cold PBS, lysed in 0.8 ml MNE buffer (25 mM MES, pH 6.5, 0.15 M NaCl, and 5 mM EDTA) containing 1% Triton X-100, fresh protease and phosphatase inhibitors, and kept on ice for 20 minutes. Lysates were then homogenized with 10 strokes of a tight fitting Dounce homogenizer and spun down at 4000g at 4°C for 10 minutes. Supernatant (0.8 ml) was mixed with the same volume of 80% sucrose prepared in MNE buffer and was placed at the bottom of an ultracentrifuge tube. Then 1.6 ml of 30% sucrose and 0.8 ml of 5% sucrose were overlaid on top of the sample to form a 5 % to 30 % discontinuous sucrose gradient. After 18 hours of centrifugation at 200,000 g and 4°C in a swinging bucket rotor, 0.4 ml samples were collected carefully from the top of each fraction. A band confined to fractions 2 through 4 was designated as caveolae/lipid raft—enriched membrane domains. 75 3.11. Isolation of Plasma Membrane Domains Cells were lysed in hypotonic buffer (10 mM HEPES, pH 7.4, 1 mM EDTA, 1 mM MgC12 freshly added with protease inhibitors) for 30 minutes on ice, then homogenized with 20 strokes in a nonstick (Teflon; DuPont, Wilmington, DE) glass homogenizer and Spun down at 500g for 5 minutes. Supematants were subjected to centrifugation (16,900g, 60 minutes, 4°C) and pellets were collected as plasma membrane—enriched fractions. 3.12. Lipid Extraction 3 x 106 HREC were washed twice with cold PBS, collected in 500 uL cold 40% methanol, and then homogenized in a glass homogenizer on ice. The cell homogenate was transferred to a screw-cap glass tube and lipids were extracted as previously described (51). The samples were dried under a stream of nitrogen and re-extracted twice to remove non-lipid contaminants. The lipid film was dried under nitrogen, then further dried overnight in a speedvac, and resuspended in an appropriate amount of isopropanol : methanol : chloroform (4 : 2 : 1) based on the protein concentration of each sample. 3.13. Mass Spectrometry Analysis Synthetic lipid standards ( Sphingolipid Mix 1, Avanti) were added to lipid extracts used in mass spectrometry analysis at 20 nmol/mg protein and 5 nmol/mg protein, respectively. Prior to analysis, lipids were appropriately diluted in isopropanol/methanol/chlorofonn (4 : 2 : 1 v /v/ v) containing 20 mM ammonium 76 hydroxide, centrifuged, then loaded into Whatrnan Multichem 96-well plates (Fisher Scientific, Pittsburgh, PA) and sealed with Teflon Ultra-Thin Sealing Tape (Analytical Sales and Services, Pompton Plains, NJ). Lipids were introduced to a Thermo model TSQ Quantum Ultra triple quadrupole mass spectrometer (San Jose, CA) as previously described (51). Ceramide (Cer) molecular species were monitored as their [M+H]+ ions by tandem mass spectrometry (MS/MS) precursor ion scanning for the formation of characteristic product ions at m/z 264.4 (Cer) in positive ionization mode. All MS/MS spectra were acquired automatically for 5-10 minutes at a rate of 500 m/z sec'l by methods created using Xcalibur software (Thermo, San Jose, CA). Correction for 13.C isotope effects and quantitation of lipid molecular species against appropriate internal standards was performed using the Lipid Mass Spectrum Analysis (LIMSA) software (52) peak model fit algorithm in conjunction with an expanded user-defined database of hypothetical lipid compounds. Ceramide molecular species were quantitated ratiometrically by comparison with the peak area of a synthetic lipid internal standard. The LIMSA software peak model fit algorithm was used as previously described for peak finding and isotope correction to arrive at an "absolute" value for each identified lipid species. All identified ceramide species were summed in order to quantify the total ceramide amount in the samples. MS/MS collision energies, CID gas pressure, and Q1 and Q3 settings were optimized using synthetic lipid standards corresponding to the individual lipid classes of interest. 77 3.14. Statistical Analysis Data are expressed as the mean i SD. Factorial ANOVA with the post hoc Tukey test (GraphPad PrismS, GraphPad Software, San Diego, CA) was used for comparing the data obtained from independent samples. Significance was established at P<0.05. 78 4. Results 4.1. Dose Response of TNFa. and IL-16 Induced CAMS Expression in HREC To determine the dose of cytokines necessary to induce an inflammatory response in HREC, dose response curves for TNFa (0-20ng/ml) and IL-IB (0.5-10ng/ml) were established using CAMS (ICAM-1 and VCAM-1) expression as a measure. The induction of ICAM-1 and VCAM-l by TNFa and IL-10 was assessed by immunoblot analysis (Figure 2.1). Based on these data, we used 10 ng/ml TNFa and 5 ng/ml IL-lB in the further studies. 4.2. TNFa. and IL-IB Induce ASMase and NSMase Activation in HREC The effect of proinflammatory cytokines TNFa and IL-1 [3 known to be increased in diabetic eyes on ASMase and NSMase activity in HREC was first determined. Cells were stimulated with either 5ng/ml lL-IB or 10 ng/ml TNFa for lSsec — 2min. The cells were then collected for ASMase and NSMase activity analysis using the Amplex Red Sphingomyelinase assay kit. IL-lB treatment increased ASMase activity in HREC as early as 15 seconds of stimulation and ASMase activity remained significantly elevated during 30 seconds of stimulation (Figure 2.2A). Similar results were obtained for NSMase activity, where IL-lB stimulation induced a maximum activation after 45 seconds of treatment (Figure 2.2 C). HREC treatment with pro-inflammatory cytokine TNFa also Significantly induced ASMase and NSMase activation after 45 seconds of stimulation (Figure 2.2, B and D). 79 4.3. Inhibition of Cytokine-Induced ASMase and NSMase Activation by DHA in HREC DHA, the most abundant w3-PUFA in the retina, has been shown to have a pronounced anti-inflammatory effect, inhibiting cytokine-induced CAMS expression in HREC and endothelial cell activation (12). To determine the effect of DHA on basal ASMase and NSMase activity, HREC were treated with DHA or linoleic acid (lipid control) for 24 hours. Pretreatment of HREC with 100 uM of BSA-bound DHA significantly decreased both ASMase (Figures 2.3 A, B, E) and NSMase (Figures 2.3 C, D, F) mRNA expression and activity level. Pretreatment of HREC with 100 uM of BSA- bound linoleic acid had no effect on ASMase or NSMase basal activity when compared to vehicle control (BSA). We next determined the effect of DHA on TNFa and IL-lB induced ASMase and NSMase activity. As shown in Figure 2.3, DHA, but not linoleic acid pretreatment of HREC, significantly down regulated TNFa and IL-lB-induced ASMase (Figure 2.3 A and B) and NSMase (Figure 2.3 C and D) activity. To validate the effect of DHA on cytokine-induced ASMase and NSMase activation, HREC were treated with 15 pM desiprarnine (53-54) and 25 uM GW4869 (55) prior to addition of stimulatory agents. Inhibition of cytokine-induced ASMase activity by desipramine is shown in Figure 2.4 A and inhibition of cytokine-induced NSMase activity by GW4869 is shown in Figure 2.4 B. 80 4.4. Effect of ASMase and NSMase Inhibition and/or Gene Silencing on TNFa and IL-IB Induced CAMS Expression To establish the role of ASMase and NSMase in cytokine-induced inflammatory signaling in HREC, ASMase and NSMase activity was inhibited using desipramine (Sigma) and GW4869 (Sigma), respectively. HREC were treated with 15 uM desipramine (53-54) for one hour and 25 uM GW4869 (55) for 30 minutes prior to TNFa (10 ng/ml) stimulation. Six hours after cytokine stimulation the activation of inflammatory pathways was analyzed using ICAM-1 and VCAM-l protein expression levels as a measure. As shown in Figure 2.5, inhibition of ASMase Significantly reduced the TNFa induced adhesion molecules expression, similarly to results achieved with DHA treatment. Inhibition of ASMase also reduced IL-1I3 induced ICAM-l expression (Figure 2.11 A, B and C). Interestingly, inhibition of NSMase did not have a significant effect on TNFa and IL-1 [3 induced ICAM-l expression, and although the effect on TNFa induced VCAM-1 expression was significant, it was less pronounced that the effect of ASMase inhibition or DHA. Gene silencing of ASMase and NSMase (100 nM siRNA for ASMase and NSMase) significantly decreased TNFa and IL-lB-induced ICAM-1 and VCAM-1 expression in HREC (Figure 2.6) as compared to untreated or control siRNA (100 nM) treated cells. Similarly to the inhibitors study, ASMase gene silencing had more pronounced inhibitory effect compared to NSMase gene silencing. No additive effect on cytokine induced CAM expression was observed in the samples where both ASMase and NSMase gene silencing was attained. 81 4.5. Inhibition of ASMase and NSMase Activity by DHA To determine the time course of DHA effect on ASMase and NSMase activity, HREC were treated with BSA-bound DHA, or BSA alone. First, the dose of DHA to be used in this experiment was selected based on dose—response experiment for ASMase (Figure 2.7 A) and NSMase (Figure 2.7 C) activity. The time course for DHA effect on ASMase and NSMase activity in HREC showed a gradual decrease in ASMase (Figure 2.7 B) and NSMase (Figure 2.7 D) activity with an early time point of 1 hour and 2 hours, respectively, and a maximum effect on both enzymes activity after 24 hours of DHA treatment. To determine whether the effect of DHA on sphingomyelinases activity is due to a direct interaction with the enzymes, we performed the sphingomyelinase assay in a cell-free system where 0.5 mU of bacterial sphingomyelinase (homolog of NSMase) was incubated with 5 to 80 uM fatty acid (DHA, linoleic acid). In this cell fi'ee system fatty acids were used without a carrier molecule. Free fatty acid concentration of 5 mM without a carrier in cell free system corresponds to 25 mM in cell culture experiments where fatty acids are complexed to BSA at 1:5 molar ratio. No fatty acids were added to the control samples. We found no direct effect of DHA or linoleic acid on SMase activity (Figure 2.8), strongly suggesting that direct binding of DHA to sphingomyelinases does not play a role in the observed inhibitory effects. 4.6. Inhibition of ASMase and NSMase Activity by MCD DHA incorporation into the fatty acyl chains of phospholipids in caveolae/lipid rafts causes cholesterol displacement from these specialized membrane microdomains 82 (47). To prove that lipid composition of caveolae/lipid rafts controls sphingomyelinase activity, we determined the effect of cholesterol depletion on ASMase and NSMase in HREC. Caveolae/lipid raft fractions were isolated by sucrose discontinuous gradient ultracentrifugation based on their insolubility in Triton-X-100 at 4°C. Caveolin-1, a caveolae marker, was used to identify the lipid raft fractions in density gradient fractions 2 to 4 (Figure 2.9 A). MCD and DHA pretreatment of HREC dramatically decreased cholesterol content of the raft fractions (Figure 2.9 B) when compared to cholesterol content of caveolae/lipid raft fractions isolated from BSA and linoleic acid pretreated HREC. Pretreatment of HREC with 8 mM MCD for 30 min decreased NSMase activity by 42%, similarly to DHA effect on NSMase activity (Figure 2.9 C). Cholesterol replenishment with 25 pM water-soluble cholesterol reversed the inhibitory effect on NSMase activity (Figure 2.9 C). NSMase activity was only assessed in the whole cell lysate since NSMase is only localized to the plasma membrane and activity in the whole cell lysate represents the quantification of NSMase activity in the plasma membrane compartment. MCD treatment of HREC had no effect on whole cell lysate ASMase activity and cholesterol replenishment did not restore ASMase activity in DHA treated HREC (Figure 2.9 D). AS a significant portion of cellular ASMase is in lysosomal compartment that would not be affected by MCD and cholesterol treatments used, we next analyzed ASMase activity in plasma membrane fiaction. ASMase activity was significantly decreased in plasma membrane fraction isolated from MCD treated HREC and 83 cholesterol replenishment in DHA treated cells restored ASMase activity in this cellular compartment (Figure 2.9 E). 4.7. No Effect of Ceramide de Nova Synthesis Inhibition on TNFa and IL-18 Induced CAMS Expression in HREC Ceramides can be synthesized in the cells at the membrane level from sphingomyelin hydrolysis by ASMase and/or NSMase or in the endoplasmic reticulum by de nova synthesis. We next determined whether ceramide de novo synthesis pathway is involved in mediating pro-inflammatory cytokine signaling in HREC. Fumonisin Blwas used to inhibit ceramide synthase, the enzyme that catalyzes the final step of de nova pathway of ceramide synthesis. Cells were treated for 16 hours with 50 uM Fumonisin B1 prior to TNFa (10 ng/ml) and IL-1 I3 (5 ng/ml) stimulation. The expression levels of ICAM-1 and VCAM-l were analyzed by western blot (Figure 2.10, A and D). Fumonisin B1 pretreatment of HREC did not affect TNFa and IL-10 induced adhesion molecules expression. In contrast, as Shown above, inhibition of ASMase significantly decreased TNFa and IL-lB induced ICAM-1 and VCAM-1 expression in HREC. These data suggest that ceramide production at the membrane level, rather than total cellular ceramide level, plays an essential role in modulating cytokine induced inflammatory signaling in HREC. Decreased ceramide levels in Fumonisin B1 treated HREC were confirmed by tandem mass spectrometry analysis. Inhibition of ceramide synthase by incubation of HREC with Fumonisin Bl resulted in a 26.55i9.45% decrease in HREC total ceramide levels, as expected (Figure 2.10 l). 84 TNFa (ng/mL) 0 1 5 10 20 ICAM-1 ...... ...-nag 49““ VCAM-1 «up ’ u _ Tubulin - — - — - lL-1B (ng/mL) 0 0.5 1 5 10 ICAM-1 ...-Eggs VCAM-1 If .q" u q Tubulin W Figure 2.1. Dose response of cytokine-induced CAMS expression in HREC. HREC maintained in serum fi'ee media overnight were stimulated with 0-20 ng/ml TNFa and 0- 10 ng/ml IL-lB for six hours. The induction of ICAM-1 and VCAM-l by TNFa (A) and IL-1 B (B) was assessed by immunoblot analysis. Equal amounts of protein were added to each lane as confirmed by tubulin level. 85 ASMase .E ,9 9 "3 a Q 4: 8 g 1 n’ I a, L .... a ‘z3 a?! E V 3:) 0,7 -- lL-1B 0 15 30 45 60 120 Time (seconds) B' ASMase 1.3 I ‘ 0.9e TNF“ o 15 30 45 60120 ASMase activity/pg protein (Normalized units) Time (seconds) Figure 2.2. Cytokine induced ASMase and NSMase activation in HREC. HREC cultured on 100m plates were stimulated with 5 ng/mL IL-l B (A,C) and 10 ng/mL TNFa (B,D) for 0-120 seconds. Each time point represents an individual plate. The cells were collected for ASMase (A,B) and NSMase (C,D) activity analysis using the Amplex Red Sphingomyelinase assay kit. IL-lB significantly induced ASMase (A) activation after 15 seconds of stimulation, with a maximal activation after 30 seconds of stimulation. TNFa induced ASMase (B) activation after 45 seconds of stimulation. 86 NSMase .s .9 3 a 3-5 * g s e 8 2.5 .2 E : '. 3 o 1.5 . ”II .3. A to Z - _.-l 2 V o E" (D 2 0.5 . , ._ w”. O 15 30 45 60 120 lL-1|3 Time (seconds) D. NSMase .5 2-5 2 * o A -P 5 £3 2 - T. c: C 3 . i --. .2 .u 1.5 - g g 1 . . . . . . $3.. *5... § 2 V " (I) TNFa 0 15 30 45 60120 Time (seconds) Figure 2.2 (continued). Cytokine induced ASMase and NSMase activation in HREC. IL-lB (C) or TNFa (D) induced maximal NSMase activation after 45 seconds of stimulation. Results are presented as mean i SD of 3 independent experiments. *P<0.05 compared to basal expression level. 87 P e: .E ASMase ASMase o m C E- 3 1.5 - 1 g 8 .5 .2 E ‘65 E 1 r 1 m E 3‘ a a * ' cu g 0.5 < 0.5 ' 2 a) ‘5 _ _O._e_~-_ .. p- 0_‘ : Stimulation - IL-1B i I BSA l [j Linoleic acidl ' i E] DHA i C. D. NSMase NSMase .E a 3 2.5 # 9 A it 0- .-‘_9 2.5 A f c) C T 2 ' 1 D s v 2 ._ cu 1.5 < .2 .5 4g E 1.5 a 1 C?) 0.5 . 0.5 ‘ z . I----z______,- . ,0,” - . - . -_ : * ,. _ , Q ‘1 Stimulation - lL-1B - Figure 2.3. Inhibition of cytokine induced ASMase and NSMase activation, as well as ASMase and NSMase expression by DHA in HREC. HREC treated with 100 pM fatty acid (DHA or linoleic acid) complexed to BSA, or BSA alone (vehicle control) overnight (24 hours) were stimulated with either 5 ng/mL lL-lB (A) for 30 seconds to assess the activation of ASMase and for 45 seconds to assess the activation of NSMase; 88 F" '51 ASMase NSMase g 0.00003 0.0003 - '5 2 0.00006 i g. i 0.0002 A 0 l 3 0.00004 3 g l 0.0001 — g 0.00002 i r— 0 . __ 0 - __ I BSA E] DHA Figure 2.3 (continued). Inhibition of cytokine induced ASMase and NSMase activation, as well as ASMase and NSMase expression by DHA in HREC. HREC treated with 100 pM fatty acid (DHA or linoleic acid) complexed to BSA, or BSA alone (vehicle control) overnight (24 hours) were stimulated with with 10 ng/mL TNFa for 45 seconds to assess both ASMase and NSMase activation. The cells were then collected for ASMase (A,B) and NSMase (C,D) activity analysis using the Amplex Red Sphingomyelinase assay kit. DHA decreased both ASMase (A,B) and NSMase (C,D) basal activity and down-regulated cytokine-induced sphingomyelinases activity. RT-PCR analysis of ASMase (E) and NSMase (F) expression in HREC with or without DHA pretreatment. Results are presented as mean i SD of 3 independent experiments. *P<0.05 compared to basal expression level in BSA and linoleic acid treated HREC, #P<0.05 compared to non-stimulated HREC, IP<0.05 compared to cytokine stimulated HREC. 89 ASMase ASMase activity/pg protein (Normalized units) BSA BSA f;.:-_Ii<'ig,g.g§-*::_.:.;‘?ng;._-1'73, DHA D ir min £3)?“ \ esp a e k B. Y IL-1B 3 _, NSMase ASMase activity/pg protein (Normalized units) IL-10 Figure 2.4. Inhibition of ASMase and NSMase cytokine-induced activation by DHA and sphingomyelinases inhibitors. HREC treated with 100 uM DHA complexed to BSA overnight and inhibitors for ASMase (Desipramine, 15 BM for 1 hour) or NSMase (GW4869, 25 BM for 30 min) down-regulated IL-lB-induced ASMase (A) and NSMase (B) activity below the basal level, respectively. Results are presented as mean 1- SD of 3 independent experiments. #P<0.05 compared to basal expression level in BSA treated HREC, 'P<0.05 compared to IL-lB stimulated, HREC. 90 BSA + + + + + + TNFa - + + + + + DHA - - + - - - G W 4869 - - - + - + Desipramine - - - - + + Cyclophilin M [CAM-1 lCAM-1/ thlaophilin (Arbitrary units) Desipramine _—4 * <1: <0 0’ (<15 :1: g 5% In 0 Eco E g? (D 9.; :00 8+ TNFa Figure 2.5. Decrease in TNFa induced CAMS expression by ASMase and NSMase inhibition in HREC. 91 VCANL1 #3: ICAM-1/ Cyclophilin (Arbitrary units) ores-crooo * ill * * (D a) 0) <75 <75 E 3 .5 58 m m D g E Egg <0 E <9 a .93 8 o f TNFa Figure 2.5 (continued). Decrease in TNFa. induced CAMs expression by ASMase and NSMase inhibition in HREC. HREC were treated with 100 uM DHA complexed to BSA, or BSA alone (vehicle control) for 24 hours, or the inhibitors for ASMase (Desipramine, 15 pM for 1 hour) or NSMase (GW4869, 25 M for 30 min) prior to 10 ng/mL TNFa stimulation. The induction of ICAM-1 and VCAM-l was assessed 6 hours later by irnmunoblot analysis (A). DHA, as well as Desipramine Significantly reduced the TNFa induced CAM expression. GW4869 significantly inhibited TNFa induced VCAM-1 expression. Quantitative analysis of the data presented in panel (A) for ICAM- 1 (B) and VCAM-l (C) expression. The results are meaniSD of 4 independent experiments. #P<0.05 compared to control cells; *P<0.05 compared to TNFa stimulated cells. 92 siRNA Control ASMase I“ M ASMase -- Tubulin siRNA Control NSMase F.” m ‘ NSMase ‘fl Tubulin Figure 2.6. Inhibition in cytokine-induced CAMS expression by ASMase and NSMase gene silencing in HREC. ASMase (A) and NSMase (B) protein level in control siRNA, ASMase siRNA and NSMase siRNA treated HREC was assessed by immunoblot analysiS.Gene silencing of ASMase and NSMase (100 nM siRNA for ASMase and NSMase) significantly decreased TNFa (C) and IL-lB (F) induced ICAM-1 and VCAM- 1 expression in HREC. In contrast, control siRNA treatment (100 nM) had no effect. Quantitative analysis of the data presented in C and F is shown in D,E and G,H respectively. Equal amounts of protein were added to each lane, as confirmed by tubulin levels. Results are presented as meaniSD of 5 independent experiments. # P<0.05 compared to control cells; *P<0.05 compared to TNFa and IL-1 B stimulated cells. 93 electroporation + + + + + + TNFa - + + + + + siRNA control - - + - .. - siRNA ASMase - - - + - + siRNA NSMase - - - - + + ICAM-1 7‘ "‘2: VCAM-1 ...... 3 - ....” .. ..., Tubulin - * _. III-fl ~ and D. [CAM-1 1.5~ ET; 3*: :53 ': z~ ‘T 9 2.1: i 0 0i i i III siRNA Control ASMase NSMase ASMase +NSMase TNFa Figure 2.6 (continued). Inhibition in cytokine-induced CAMS expression by ASMase and NSMase gene silencing in HREC. 94 VCA M-1 * VCAM-1/ Tubulin (Arbitrary units) . I 4—0 SIRNA g ‘2“ 2 O a) (£3 < + TNFa F. electroporation + + + + + + 151,3 - + + + + + siRNA control - - + - - - siRNA ASMase - - - + - + - - - - + + siRNA NSMase ICAM'1 ‘ I ~ ‘ my old ......" VCAM-1 { on. .0 w-v- but... «.14.... TUbulin W Figure 2.6 (continued). Inhibition in cytokine-induced CAMs expression by ASMase and NSMase gene silencing in HREC. 95 [CAM-1 1.5- =2 :5 # # .o c 1 .3 3 u. ?E * T * E 1‘: 5510-5 0 6 (Did); 0% . I I ,1: (D 0) mm SIRNA g g g g: a) 0 ‘2 ‘2 2’2; lL-1B H. VCAM-1 1.5 E 13 3 4: .135 1 : E I E * E 20.5a o < > V NSMase ASMase +NSMase lL-1 E Figure 2.6 (continued). Inhibition in cytokine-induced CAMS expression by ASMase and NSMase gene silencing in HREC. 96 .3’ cu ASMase . ASMase e111. 1 000 50° *"'§°---i--“§'"* * 51* o§§§ ASMase activity/pg protein (Normalized units) Li... ...} * °°"§ BSA 50 75 100 - 2’ 5’ 10’ 1h 2h 4h 6h12h 24h DHA (nM) DHA treatment C. D NSMase NSMase C '6 ‘9' A 600 2, ‘3 40° E .. ..f.. g: 3 400 "t. °§....l. * i g 0N, 2m * i... '%'-f * '- = ‘ .’ ' .. T g g m 200 l * m 0 "mi as Z 2 V 0 - o . , (g BSA 50 75 100 - 2’ 5’ 10’ 1h 2h 4h 6h 12h 24h DHA (nM) DHA treatment Figure 2.7. Inhibition of ASMase and NSMase activity by DHA. HREC treated with 100 M DHA complexed to BSA, or BSA vehicle control overnight (24 hours) were collected for ASMase (A) and NSMase (D) activity analysis using the Amplex Red Sphingomyelinase assay kit. The optimal dose of DHA to inhibit ASMase (A) and NSMase (C) activity in HREC was 100 1.1M. The time course for DHA effect on ASMase and NSMase activity in HREC showed a gradual decrease in ASMase (B) and NSMase (D) activity with an early time point of 1 hour and 2 hours, respectively and a maximum effect on both enzymes activity afier 24 hours of DHA treatment. The results are meaniSD of 3 independent experiments. 'P<0.05 compared to BSA vehicle control. 97 1-4 1.2 .+. 99 moo SMase actIVIty (Arbitrary units) 0.4 0.2 SuM IOpM ZOuM 40uM 80uM Fatty acid concentration ; I Control [:1 DHA | { [:] Linoleic j Figure 2.8. No direct effect of DHA on sphingomyelinase (SMase) activity in a cell- free system. To determine whether DHA decreases SMase activity through direct interaction with the enzyme, we measured SMase activity in a cell free system, where 0.5 mU of bacterial SMase (provided in the Amplex Red Sphingomyelinase assay kit) were incubated with 5 to 80 uM fatty acids (DHA, linoleic acid). A concentration of 20 “M free fatty acid corresponds to a concentration of 100 pM fatty acid complexed to BSA in 5:1 molar ratio. No fatty acids were used for the control wells. The assay was performed as described in the Methods. No effect of DHA or linoleic acid on SMase activity was observed when compared to control. 98 Fractions :3...‘ ' “- Linoleic acid .: ...... DHA .. ._ ~-~ MCD a... «1 BSA Caveolin-1 B. Cholesterol 200 150 m (Arbitrary units) 1oo , * 50 I m o 2 _ BSALinoleic DHA MCD Cholesterol level/ pg of protein Figure 2.9. Effect of cholesterol depletion on ASMase and NSMase activity in HREC. Caveolae/lipid raft fractions were isolated by sucrose discontinuous gradient ultracentrifugation based on their insolubility in Triton-X-IOO at 4°C and identified by western blotting as fractions 2-4 based on the caveolin-1 distribution (A). MCD (white bars) and DHA (dark gray bars) pretreatment of HREC significantly decreased cholesterol content of the caveolae fractions when compared to BSA (carrier control) and linoleic acid (lipid control) (B). 99 Figure 2.9 (continued). Effect of cholesterol depletion on ASMase and NSMase activity in HREC. NSMase activity was inhibited by cholesterol depletion caused by the treatment with 100 uM DHA overnight (gray bar) or 8 mM MCD for 30 min (white bar)(C). The inhibitory effect of DHA on NSMase activity was reversed by cholesterol replenishment using 25 14M water-soluble cholesterol for 30 minutes (hatched gray bar, C). The inhibitory effect of DHA on ASMase activity was not mimicked by MCD and cholesterol replenishment did not restore ASMase activity in DHA treated HREC in whole cell lysates (D). NSMase activity/pg protein ASMase activity/pg protein (Normalized units) (Normalized units) NSMase activity Whole cell lysate 400 7 l 300 4' . l :2. . 200 t 1oo | $1.}? 0 “/29“ , , BSA DHA DHA MCD +cholesterol ASMase activity Whole cell lysate . Jr 300 i 200 1; 100 BSA DHA DHA MCD +cholestero| 100 ASMase activity Plasma membrane P1 BSA DHA DHA MCD ASMase activity/pg protein (Normalized Ul‘lltS) +cholesterol Figure 2.9 (continued). Effect of cholesterol depletion on ASMase and NSMase activity in HREC. However, cholesterol depletion inhibited ASMase activity and cholesterol replenishment reversed the inhibitory effect of DHA on ASMase activity in the plasma membrane fraction (E). Results are presented as meaniSD of 3 independent experiments. *P<0.05 compared to BSA control. #P<0.05 compared to DHA. 101 IL-l B - + + + Fumonisin Bl - - + - Desipramine - - - + ICAM-1 '“'“ _Q-— VCAM-1 . ~ ~ ...... Tubulin M B. C. ICAM-1 VCAM-1 c A15 ~ # # 1,5 a .2 # g S 1 . 1‘. # E s g g 0.5 I 0.5 _, * . o 1 a a _ 01 I I E .93) I . E .E .E E .E E a g a g 8 a g a E: 0 ..t 0 lL-1B IL-1t3 Figure 2.10. No effect of de novo ceramide production pathway inhibition on cytokine induced CAMs expression in HREC. Fumonisin B1 was used to inhibit ceramide synthase, the enzyme that catalyzes the final step in the de novo pathway of ceramide synthesis. Cells were treated for 16 hours with 50 uM Fumonisin Bl prior to TNFa (10 ng/ml) and IL-IB (5 ng/ml) stimulation. The expression levels of ICAM-1 and VCAM-l were analyzed by western blot. 102 TNFa - + + + Fumonisin Bl - - + _ Desipramine - - _ + ICAM-1 1. g. ‘ I a - VCAM-1 .. a- mi: - .. * Tubulin - - - - [CAM-1 VCAM-1 1.5 0'5 0.5 o 1 Protem/ Tubulin (Arbitrary units) O i Desipramine -—-t . I I f E | I a g E .E E .2 .2 9 c c Q 0 o .- g g 3 LL “- 0 TN Fa TN Fa Figure 2.10 (continued). No effect of de novo ceramide production pathway inhibition on cytokine induced CAMs expression in HREC. Representative blots are shown in A and D, and quantitative analysis of 4 independent experiments is shown in B,C,E and F. Fumonisin Bl pre-treatment of HREC did not affect TNFa and IL-lB induced adhesion molecule expression. In contrast, inhibition of ASMase significantly decreased TNFa and IL-lB induced ICAM-1 and VCAM-1 expression. 103 Control HREC C"3T(c11t3:1r24:0) " Cer . . 650 °\o (d18.1116.0) Cer _ _ V - (d18-1l24.1) Cer - . 8IOO 0:1“: 1,120) 538 6 48‘»... |Satan/25.0) 53 «(432) Cer ‘ _Cer(d18:1/22:0) 6(64) 5 (d18.1118.0) 622 g L 566 < .__A x 11 H. Fumonisin 81 treated HREC cer(d*11;1I24:0) A German/16:0) 650 Ce 5 ,Cefidtsmzn) 538 Ceqmmm.” “(a r(d18:1l25.0) 8100 (LS) 6481 '(l.S.) g 432 C9r(d18:1t22:0) “ 664 1:7 “Rattan/18:0) 622 3 566 .0 < ___r___________, 470 570 670 mlz Figure 2.10 (continued). No effect of de novo ceramide production pathway inhibition on cytokine induced CAMs expression in HREC. Ceramide levels determined by tandem mass spectrometry precursor ion mode scanning for the characteristic ceramide product ion at m/z 264.4 in control and Fumonisin Bl treated HREC are presented in G and H respectively. 104 1 400 1 200 1 000 Ceramide level (pmol/mg protein) I Control El Fumonisin B1 Figure 2.10 (continued). No effect of de nova ceramide production pathway inhibition on cytokine induced CAMs expression in HREC. Quantification of ceramide levels presented in 1 demonstrates significant decrease in ceramide levels in Fumonisin B1 treated compared to control HREC. Results are presented as meaniSD of 3 independent experiments. *P<0.05 compared to cytokine stimulated level. #P<0.05 compared to control. 105 5. Discussion Omega 3 PUFAS have been long accepted to modulate inflammatory processes and are now widely used in the clinic as an adjuvant immunosuppressant in the treatment of various diseases with an inflammatory component (56). In a recent article, Connor et al. showed that increasing omega 3 PUF A retinal levels by diet or genetic means may be of benefit in preventing retinopathy and the DHA protective effect was mediated, in part, by suppression of retinal TNFa gene expression and protein level in a mouse model of oxygen-induced retinopathy (5). We have previously shown that DHA suppresses cytokine induced adhesion molecule expression in endothelial cells through cholesterol depletion and displacement of important signaling molecules from caveolae/lipid rafts (47). We now demonstrate that DHA treated HREC exhibit decreased basal and TNFa and IL-IB induced activity of ASMase and NSMase enzymes. The time course showed maximum effect of DHA on sphingomyelinase activity after more than 12 hours of treatment. This effect can be explained by our previously published observation that DHA incorporates into caveolae/lipid rafts phospholipids at a rate of 10% in 1.5 hours and 90% in 24 hours and dramatically alters the lipid environment of these specialized microdomains, decreasing their cholesterol content by approximately 70% (47). Both ASMase and NSMase have been described to localize in the caveolae/lipid raft compartment of various cell types. Although ASMase was initially described as a strictly lysosomal enzyme because of its optimum activity at pH 4.5-5.0, Liu and Anderson have described the plasma membrane form of ASMase in caveolae and they have shown that IL-IB stimulates its activity in this compartment (27). Veldman et al. 106 described a NSMase in caveolae isolated from human skin fibroblasts where the enzyme appeared to interact with the scaffolding domain of caveolin protein (57). NSMase has two putative transmembrane domains and five palmitoylation sites (29) and thus likely has affinity for caveolae/lipid rafts microdomains. Cholesterol depletion as well as displacement of signaling molecules essential for sphingomyelinases activation from caveolae/lipid rafts by DHA may be the mechanism of DHA inhibitory effect on ASMase and NSMase activity. Indeed, in this study, cholesterol depletion using MCD decreased NSMase activity to the level similar to that observed with DHA. In contrast to NSMase, ASMase has a major intracellular lysosomal component. The function and control of ASMase activity in the plasma membrane and in lysosomal compartments are likely to be different. Thus, it is not surprising that whole cell ASMase activity, which represents lysosomal and plasma membrane ASMase combined, was not affected by short term MCD and cholesterol treatments the same way as NSMase, which has only plasma membrane localization. Plasma membrane ASMase activity was however, significantly decreased by MCD, and cholesterol replenishment in DHA treated cells restored ASMase activity in the plasma membrane. Pro-inflammatory cytokines TNFa and IL-IB have been shown to activate cellular ASMase in various cell types (27, 58-60) and our results are in agreement with these studies. We have found that IL-IB treatment of HREC induced rapid activation of ASMase afier only 15 seconds of stimulation, while TNFa increased both ASMase and NSMase activity after 45 seconds of stimulation. This rapid and transient time course of the effect is in agreement with the fact that induction of the inflammatory response is always transient and the extent of the initial event is always small as it is later amplified 107 several fold with each signal transduction step. For instance, as demonstrated in numerous studies of the NF KB pathway, IKBa is phosphorylated within a minute from cytokine stimulation and is dephosphorylated back to the control state by 5 min; in GPCR signaling ERK phosphorylation occurs within 30 seconds of receptor activation and it is back to normal level by 2 minutes. Indeed, a recent study described the importance of transient versus sustained ERK phosphorylation in inducing different signaling pathways (61). Moreover, sphingomyelinase literature is replete with demonstration of the very fast (within a minute) and transient nature of sphingomyelinase activation, and the fact that it is so fast is often cited to promote the notion that sphingomyelinase is an important first responder (58-59, 62). Although the induction of sphingomyelinases is transient, the effect of the sphingomyelinase activation, e.g., conversion of sphingomyelin to ceramide and further activation of the NFKB pathway and inflammatory genes, such as ICAM-1 and VCAM-l, is long lasting. Indeed, in this study inhibition and/or gene silencing of ASMase and NSMase decreased pro-inflammatory cytokine TNFa and IL-lB induced adhesion molecules expression in HREC. The majority of studies on sphingomyelinase activation have used as a model system the p55 TNFa receptor, showing that this receptor has a modular structure, with particular domains for different sphingomyelinases. The intracellular C-terminal part of the p55 receptor, which contains the death domain (DD), appears to be necessary for ASMase activation; the exact molecular mechanism is not known, but Schwandner et al. have reported ASMase activation (seconds to minutes) in response to TNFa through overexpression of TRADD (TNF receptor associated DD) and/or F ADD (Pas-associating protein with DD) (63). Kronke et al. have defined a region of this receptor that is adjacent 108 to the death domain and is required for NSMase activation (64). This motif was named the NSMase activation domain. The same authors have also described a novel protein (factor associated with NSMase activation, FAN) that binds the NSMase activation domain on the TNFa receptor and is required for NSMase activation (65). The mechanism by which lL-lB stimulates ASMase and NSMase activity is not yet elucidated. Inhibition and/or gene silencing of ASMase and NSMase decreased pro- inflammatory cytokine TNFa and IL-18 induced adhesion molecules expression in HREC. Both ASMase and NSMase are specific enzymes with phospholipase C activity, which hydrolyze the sphingomyelin phosphodiester bond to produce ceramide and phosphocholine. The ability of ceramide to self-aggregate and their fusagenic properties may mediate membrane microdomain reorganization into large platforms required for receptor clustering (66-68). As TNFa and IL-IB have receptors that appear to be localized in the caveolae/lipid rafts fractions (58, 68) and protein oligomerization is an almost universal requirement for transmembrane signal transmission, release of ceramides alters the dynamics of these rafts and may drive and amplify inflammatory signal transduction processes. Moreover, there is evidence that stimulation of the sphingomyelin pathway by TNFa with sphingomyelinases-induced membrane ceramide leads to activation of nuclear factor KB (NF-KB) and marked increase in nuclear NF-KB binding in human leukemia (HL-60) cells (59). Similarly, activation of sphingomyelinases is shown to be an important signaling system for lL—lB in the murine T helper cell, EL-4 (58), and IL-18 109 action in these cells is mediated through NF-KB activation (69). NF-KB is a major transcription factor controlling the expression of an array of inflammatory response genes including adhesion molecules (70). There are two different pathways of ceramide generation in cells. The first takes place at the membrane level by hydrolysis of sphingomyelin by sphingomyelinases; the second is localized to the endoplasmic reticulum by de novo synthesis involving the enzyme ceramide synthase. The results of this study demonstrate that plasma membrane ceramide production by sphingomyelinases rather than de novo synthesis is important for inflammatory signaling in HREC. Several recent studies demonstrated that oxidized products of DHA formed either through lypoxygenase (5, 71-72), cyclooxygenase (72-73) or non-enzymatic pathways (74) possess potent anti-inflammatory properties. Although anti-inflammatory mediators formed by DHA oxidation are not likely to explain the results of this study, they might represent an important component of anti-inflammatory action of DHA in other cell types or under different experimental conditions. Endothelial cells are rich in caveolae and ASMase is preferentially expressed in endothelia (75), thus our findings may represent an identification of an endothelial-specific anti-inflammatory mechanism of DHA action. In conclusion, this study provides a novel mechanism for the anti-inflammatory effect of DHA in the primary tissue affected by retinopathy, human retina] endothelial cells. We demonstrated that DHA-induced cholesterol depletion from membrane microdomain leads to inhibition of ASMase and NSMase activity. Sphingomyelinase downregulation by inhibitors and/or gene silencing recapitulated the DHA protective 110 effect against cytokine-induced inflammation in HREC. Importantly, preclusion of cytokine-induced adhesion molecules in HREC was more pronounced in HREC when ASMase rather than NSMase inhibition/gene silencing was attained, supporting the general hypothesis that ASMase may play a more important role in active reorganization of plasma membrane microdomains and receptor clustering (28). Furthermore, Veldman et al. showed that after TNF stimulation, NSMase exited the caveolae and was followed by a significant increase in ceramide content in this compartment, suggesting that ASMase might mediate sphingomyelin hydrolysis (57). Grassme et al. demonstrated that cells derived from Niemann-Pick disease patients (with ASMase deficiency) were defective in receptor clustering and signal transduction in response to Fas, underlining the importance of ASMase in oligomerization of cell surface proteins and transmembrane signal transmission. Moreover, vascular endothelium is well recognized to express 20 times as much ASMase as any other cell type in the body (76) and this large excess of ASMase in endothelium may be involved in tissue remodeling and wound repair (77), processes that normally require waves of endothelial cell apoptosis and tissue neovascularization (78). Therefore, in the next chapter we characterized ASMase expression and cellular localization in HREC when compared to other retinal tissues affected by diabetic retinopathy, such as human retinal pigment epithelial cells and human Muller cells. Moreover, we tested the DHA effect on caveolar versus intracellular ASMase expression and we analyzed the sphingolipid composition of caveolae/lipid microdomains in control, DHA treated and/or cytokine stimulated HREC. 111 6. References 10. Lecomte, M., Paget, C., Ruggiero, D., Wiemsperger, N., and Lagarde, M. 1996. Docosahexaenoic acid is a major n-3 polyunsaturated fatty acid in bovine retinal microvessels. J Neuroch em 66:2160-2167. SanGiovanni, J.P., Chew, E.Y., Clemons, T.E., Davis, M.D., Ferris, F.L., 3rd, Gensler, G.R., Kurinij, N., Lindblad, A.S., Milton, R.C., Seddon, J.M., et a1. 2007. The relationship of dietary lipid intake and age-related macular degeneration in a case-control study: AREDS Report No. 20. Arch Ophthalmol 125:671-679. Decsi, T., Minda, H., Hermann, R., Kozari, A., Erhardt, E., Burns, 1., Molnar, S., and Soltesz, G. 2002. Polyunsaturated fatty acids in plasma and erythrocyte membrane lipids of diabetic children. Prostaglandins Leukot Essent Fatty Acids 67:203-210. Futterrnan, S., and Kupfer, C. 1968. The fatty acid composition of the retinal vasculature of normal and diabetic human eyes. Invest Ophthalmol 7:105-108. Connor, K.M., SanGiovanni, J.P., Lofqvist, C., Aderman, C.M., Chen, J., Higuchi, A., Hong, S., Pravda, E.A., Majchrzak, S., Carper, D., et a1. 2007. Increased dietary intake of omega-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nat Med 13 :868-873. McLeod, D.S., Lefer, D.J., Merges, C., and Lutty, GA. 1995. Enhanced expression of intracellular adhesion molecule-1 and P-selectin in the diabetic human retina and choroid. Am J Pathol 147:642-653. Joussen, A.M., Murata, T., Tsujikawa, A., Kirchhof, B., Bursell, S.E., and Adamis, AP. 2001. Leukocyte-mediated endothelial cell injury and death in the diabetic retina. Am J Pathol 158:147-152. Adamis, AP. 2002. Is diabetic retinopathy an inflammatory disease? Br J Ophthalmol 86:363-365. Limb, G.A., Chignell, A.H., Green, W., LeRoy, F., and Dumonde, DC. 1996. Distribution of TNF alpha and its reactive vascular adhesion molecules in fibrovascular membranes of proliferative diabetic retinopathy. Br J Ophthalmol 80:168-173. Joussen, A.M., Poulaki, V., Le, M.L., Koizumi, K., Esser, C., Janicki, H., Schraermeyer, U., Kociok, N., Fauser, S., Kirchhof, B., et al. 2004. A central role for inflammation in the pathogenesis of diabetic retinopathy. Faseb J 18:1450- 1452. 112 ll. 12. 13. 14. 15. l6. 17. 18. 19. 20. 21. Adamis, A.P., Miller, J.W., Bemal, M.T., D'Amico, D.J., Folkman, J., Yeo, T.K., and Yeo, K.T. 1994. Increased vascular endothelial grth factor levels in the vitreous of eyes with proliferative diabetic retinopathy. Am J Ophthalmol 118:445-450. Chen, W., Esselman, W.J., Jump, DB, and Busik, J.V. 2005. Anti-inflammatory effect of docosahexaenoic acid on cytokine-induced adhesion molecule expression in human retinal vascular endothelial cells. Invest Ophthalmol Vis Sci 46:4342-4347. Miyawaki-Shimizu, K., Predescu, D., Shimizu, J., Broman, M., Predescu, S., and Malik, AB. 2006. siRNA-induced caveolin-1 knockdown in mice increases lung vascular permeability via the junctional pathway. Am J Physiol Lung Cell Mol Physiol 290:L405-413. Li, S., Song, KS, and Lisanti, M.F. 1996. Expression and characterization of recombinant caveolin. Purification by polyhistidine tagging and cholesterol- dependent incorporation into defined lipid membranes. J Biol Chem 271:568-573. Murata, M., Peranen, J., Schreiner, R., Wieland, F ., Kurzchalia, T.V., and Simons, K. 1995. VIP21/caveolin is a cholesterol-binding protein. Proc Natl Acad Sci U S A 92:10339-10343. Anderson, R.G. 1993. Caveolae: where incoming and outgoing messengers meet. Proc Natl Acad Sci U S A 90: 10909-10913. Lisanti, M.P., Tang, Z., Scherer, P.B., Kubler, E., Koleske, A.J., and Sargiacomo, M. 1995. Caveolae, transmembrane signalling and cellular transformation. Mol Membr Biol 12:121-124. Anderson, R.G., and Jacobson, K. 2002. A role for lipid shells in targeting proteins to caveolae, rafts, and other lipid domains. Science 296: 1821-1825. Razani, B., and Lisanti, M.F. 2001. Caveolins and caveolae: molecular and functional relationships. Exp Cell Res 271 :36—44. Smart, B.J., Graf, G.A., McNiven, M.A., Sessa, W.C., Engelman, J.A., Scherer, P.B., Okamoto, T., and Lisanti, MP. 1999. Caveolins, liquid-ordered domains, and signal transduction. Mol Cell Biol 19:7289-7304. Sotgia, F., Razani, B., Bonuccelli, G., Schubert, W., Battista, M., Lee, H., Capozza, F., Schubert, A.L., Minetti, C., Buckley, J.T., et al. 2002. Intracellular retention of glycosylphosphatidyl inositol-linked proteins in caveolin-deficient cells. Mol Cell Biol 22:3905-3926. 113 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. Glenney, J.R., Jr. 1989. Tyrosine phosphorylation of a 22-kDa protein is correlated with transformation by Rous sarcoma virus. J Biol Chem 264:20163- 20166. Edidin, M. 2003. The state of lipid rafts: from model membranes to cells. Annu Rev Biophys Biomol Struct 32:257-283. Mathias, S., Pena, L.A., and Kolesnick, RN. 1998. Signal transduction of stress via ceramide. Biochem J 335 ( Pt 3):465-480. Bilderback, T.R., Gazula, V.R., Lisanti, M.P., and Dobrowsky, RT. 1999. Caveolin interacts with Trk A and p75(NTR) and regulates neurotrophin signaling pathways. J Biol Chem 274:257-263. Cremesti, A., Paris, F., Grassme, H., Holler, N., Tschopp, J., Fuks, Z., Gulbins, E., and Kolesnick, R. 2001. Ceramide enables fas to cap and kill. J Biol Chem 276:23954-23961. Liu, P., and Anderson, R.G. 1995. Compartmentalized production of ceramide at the cell surface. J Biol Chem 270:27179-27185. Marchesini, N., and Hannun, Y.A. 2004. Acid and neutral sphingomyelinases: roles and mechanisms of regulation. Biochem Cell Biol 82:27-44. Tani, M., and Hannun, Y.A. 2007. Neutral sphingomyelinase 2 is palmitoylated on multiple cysteine residues. Role of palmitoylation in subcellular localization. J Biol Chem 282:10047-10056. Tani, M., Ito, M., and Igarashi, Y. 2007. Ceramide/sphingosine/sphingosine 1- phosphate metabolism on the cell surface and in the extracellular space. Cell Signal 19:229-237. Grassme, H., Schwarz, H., and Gulbins, E. 2001. Molecular mechanisms of ceramide-mediated CD95 clustering. Biochem Biophys Res Commun 284:1016- 1030. Schissel, S.L., Jiang, X., Tweedie-Hardman, J ., Jeong, T., Camejo, E.H., Najib, J ., Rapp, J.H., Williams, K.J., and Tabas, I. 1998. Secretory sphingomyelinase, a product of the acid sphingomyelinase gene, can hydrolyze atherogenic lipoproteins at neutral pH. Implications for atherosclerotic lesion development. J Biol Chem 273:2738-2746. Smith, E.L., and Schuchman, EH. 2008. The unexpected role of acid sphingomyelinase in cell death and the pathophysiology of common diseases. Faseb J 22:3419-3431. 114 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. Kolesnick, R. 2002. The therapeutic potential of modulating the ceramide/sphingomyelin pathway. J Clin Invest 110:3-8. Pierini, L.M., and Maxfield, FR. 2001. Flotillas of lipid rafts fore and aft. Proc Natl Acad Sci U S A 98:9471-9473. Shaul, P.W., and Anderson, R.G. 1998. Role of plasmalemmal caveolae in signal transduction. Am J Physiol 275:L843-851. Simons, K., and Ikonen, E. 1997. Functional rafts in cell membranes. Nature 387:569-572. Grassme, H., Riethmuller, J., and Gulbins, E. 2007. Biological aspects of ceramide-enriched membrane domains. Prog Lipid Res 46: 161-1 70. Holopainen, J .M., Subramanian, M., and Kinnunen, PK. 1998. Sphingomyelinase induces lipid microdomain formation in a fluid phosphatidylcholine/sphingomyelin membrane. Biochemistry 37:17562-17570. Kolesnick, R.N., Goni, F.M., and Alonso, A. 2000. Compartmentalization of ceramide signaling: physical foundations and biological effects. J Cell Physiol 184:285-300. Nurminen, T.A., Holopainen, J .M., Zhao, H., and Kinnunen, PK. 2002. Observation of topical catalysis by sphingomyelinase coupled to microspheres. J Am Chem Soc 124:12129-12134. Bertrand, D. 1997. [Mental health and cultural issues: the return of Khmers from France to Cambodia for healing purposes]. Sante 7:330-334. Monks, C.R., Freiberg, B.A., Kupfer, H., Sciaky, N., and Kupfer, A. 1998. Three- dimensional segregation of supramolecular activation clusters in T cells. Nature 395:82-86. Ko, Y.G., Lee, J.S., Kang, Y.S., Ahn, J.H., and Seo, J.S. 1999. TNF-alpha- mediated apoptosis is initiated in caveolae-like domains. J Immunol 162:7217- 7223. Martin, M.U., and Wesche, H. 2002. Summary and comparison of the signaling mechanisms of the Toll/interleukin-l receptor family. Biochim Biophys Acta 1592:265-280. Busik, J .V., Mohr, S., and Grant, MB. 2008. Hyperglycemia-induced reactive oxygen species toxicity to endothelial cells is dependent on paracrine mediators. Diabetes 57:1952-1965. 115 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. Chen, W., Jump, D.B., Esselman, W.J., and Busik, J.V. 2007. Inhibition of cytokine signaling in human retina] endothelial cells through modification of caveolae/lipid rafts by docosahexaenoic acid. Invest Ophthalmol Vis Sci 48: 1 8-26. Jump, D.B., Clarke, S.D., MacDougald, 0., and Thelen, A. 1993. Polyunsaturated fatty acids inhibit Sl4 gene transcription in rat liver and cultured hepatocytes. Proc Natl Acad Sci U S A 90:8454-8458. Botolin, D., and Jump, DB. 2003. Selective proteolytic processing of rat hepatic sterol regulatory element binding protein-l (SREBP-l) and SREBP-Z during postnatal development. J Biol Chem 278:6959-6962. Agardh, E., Gustavsson, C., Hagert, P., Nilsson, M., and Agardh, CD. 2006. Modifying a standard method allows simultaneous extraction of RNA and protein, enabling detection of enzymes in the rat retina with low expressions and protein levels. Metabolism 55:168-174. Lydic, T.A., Busik, J .V., Esselman, W.J., and Reid, GE. 2009. Complementary precursor ion and neutral loss scan mode tandem mass spectrometry for the analysis of glycerophosphatidylethanolamine lipids fiom whole rat retina. Anal Bioanal Chem. Haimi, P., Uphoff, A., Hermansson, M., and Somerharju, P. 2006. Software tools for analysis of mass spectrometric lipidome data. Anal Chem 7 818324-833 1. Heinrich, M., Wickel, M., Schneider-Brachert, W., Sandberg, C., Gahr, J., Schwandner, R., Weber, T., Saftig, P., Peters, G, Brunner, J., et al. 1999. Cathepsin D targeted by acid sphingomyelinase-derived ceramide. Embo J 18:5252-5263. Kolzer, M., Werth, N., and Sandhoff, K. 2004. Interactions of acid sphingomyelinase and lipid bilayers in the presence of the tricyclic antidepressant desipramine. F EBS Lett 559296-98. Marchesini, N., Luberto, C., and Hannun, Y.A. 2003. Biochemical properties of mammalian neutral sphingomyelinase 2 and its role in sphingolipid metabolism. J Biol Chem 278:13775-13783. Calder, P.C. 2006. n-3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr 83:1505S-1519S. Veldman, R.J., Maestre, N., Aduib, G.M., Medin, J .A., Salvayre, R., and Levade, T. 2001. A neutral sphingomyelinase resides in sphingolipid-enriched microdomains and is inhibited by the caveolin-scaffolding domain: potential implications in tumour necrosis factor signalling. Biochem J 355:859-868. 116 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. Mathias, 8., Younes, A., Kan, C.C., Orlow, 1., Joseph, C., and Kolesnick, RN. 1993. Activation of the sphingomyelin signaling pathway in intact EL4 cells and in a cell-free system by IL-1 beta. Science 259:519-522. Yang, Z., Costanzo, M., Golde, D.W., and Kolesnick, RN. 1993. Tumor necrosis factor activation of the sphingomyelin pathway signals nuclear factor kappa B translocation in intact HL-60 cells. J Biol Chem 268:20520-20523. Marathe, S., Schissel, S.L., Yellin, M.J., Beatini, N., Mintzer, R., Williams, K.J., and Tabas, I. 1998. Human vascular endothelial cells are a rich and regulatable source of secretory sphingomyelinase. Implications for early atherogenesis and ceramide-mediated cell signaling. J Biol Chem 273:4081-4088. York, R.D., Yao, H., Dillon, T., Ellig, C.L., Eckert, S.P., McCleskey, E.W., and Stork, P.J. 1998. Rapl mediates sustained MAP kinase activation induced by nerve growth factor. Nature 392:622-626. Dressler, K.A., Mathias, S., and Kolesnick, RN. 1992. Tumor necrosis factor- alpha activates the sphingomyelin signal transduction pathway in a cell-free system. Science 255:1715-1718. Schwandner, R., Wiegmann, K., Bernardo, K., Kreder, D., and Kronke, M. 1998. TNF receptor death domain-associated proteins TRADD and FADD signal activation of acid sphingomyelinase. J Biol Chem 273:5916-5922. Wiegmann, K., Schutze, S., Machleidt, T., Witte, D., and Kronke, M. 1994. Functional dichotomy of neutral and acidic sphingomyelinases in tumor necrosis factor signaling. Cell 78: 1005-1015. Adam-Klages, 8., Adam, D., Wiegmann, K., Struve, S., Kolanus, W., Schneider- Mergener, J ., and Kronke, M. 1996. FAN, a novel WD-repeat protein, couples the p55 TNF-receptor to neutral sphingomyelinase. Cell 86:937-947. Belka, C., Marini, P., Budach, W., Schulze-Osthoff, K., Lang, F., Gulbins, E., and Barnberg, M. 1998. Radiation-induced apoptosis in human lymphocytes and lymphoma cells critically relies on the up-regulation of CD95/Fas/APO-l ligand. Radiat Res 149:588-595. Boniface, J .J ., Rabinowitz, J .D., Wulfing, C., Hampl, J ., Reich, Z., Altman, J .D., Kantor, R.M., Beeson, C., McConnell, H.M., and Davis, M.M. 1998. Initiation of signal transduction through the T cell receptor requires the multivalent engagement of peptide/MHC ligands [corrected]. Immunity 9:459-466. Natoli, G., Costanzo, A., Guido, F., Moretti, F., and Levrero, M. 1998. Apoptotic, non-apoptotic, and anti-apoptotic pathways of tumor necrosis factor signalling. Biochem Pharmacol 56:915-920. 117 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. Stylianou, E., O'Neill, L.A., Rawlinson, L., Edbrooke, M.R., Woo, P., and Saklatvala, J. 1992. Interleukin 1 induces NF-kappa B through its type I but not its type II receptor in lymphocytes. J Biol Chem 267: 15836-1 5841. Monaco, C., and Paleolog, E. 2004. Nuclear factor kappaB: a potential therapeutic target in atherosclerosis and thrombosis. Cardiovasc Res 61 :671-682. Marcheselli, V.L., Hong, S., Lukiw, W.J., Tian, X.H., Gronert, K., Musto, A., Hardy, M., Gimenez, J.M., Chiang, N., Serhan, C.N., et al. 2003. Novel docosanoids inhibit brain ischemia-reperfusion-mediated leukocyte infiltration and pro-inflammatory gene expression. J Biol Chem 278143 807-43817. Schwab, J.M., Chiang, N., Arita, M., and Serhan, ON. 2007. Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature 447:869-874. Serhan, C.N., Hong, S., Gronert, K., Colgan, S.P., Devchand, P.R., Mirick, G., and Moussignac, R.L. 2002. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J Exp Med 196:1025-1037. Musiek, E.S., Brooks, J.D., Joo, M., Brunoldi, E., Porta, A., Zanoni, G., Vidari, G., Blackwell, T.S., Montine, T.J., Milne, G.L., et al. 2008. Electrophilic Cyclopentenone Neuroprostanes Are Anti-inflammatory Mediators Formed fiom the Peroxidation of the {omega}-3 Polyunsaturated Fatty Acid Docosahexaenoic Acid. J Biol Chem 283:19927-19935. Kolesnick, R., and Fuks, Z. 2003. Radiation and ceramide-induced apoptosis. Oncogene 22:5897-5906. Garcia-Barros, M., Paris, F ., Cordon-Cardo, C., Lyden, D., Rafii, S., Haimovitz- Friedman, A., Fuks, Z., and Kolesnick, R. 2003. Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science 300:1 155-1159. Jensen, J .M., Schutze, S., Forl, M., Kronke, M., and Proksch, E. 1999. Roles for tumor necrosis factor receptor p55 and sphingomyelinase in repairing the cutaneous permeability barrier. J Clin Invest 104:1761-1770. Chavakis, E., and Dimmeler, S. 2002. Regulation of endothelial cell survival and apoptosis during angiogenesis. Arterioscler T hromb Vasc Biol 22:887-893. 118 Chapter III Docosahexaenoic Acid Regulates Caveolar Acid Sphingomyelinase Expression and Promotes Sphingomyelin Enrichment of Membrane Microdomains in Human Retinal Endothelial Cells 1. Abstract Retinal endothelial cells represent the resident vasculature affected by diabetic retinopathy. In addition to the retinal vasculature, diabetes may also result in damage to nonvascular retinal tissue; yet microangiopathy represents the key pathologic feature of diabetic retinopathy. As demonstrated in the previous chapter, acid sphingomyelinase (ASMase) represents a key molecule in the cytokine mediated cellular inflammatory cascade. To test the hypothesis that docosahexaenoic acid (DHA) has an anti-inflammatory effect in diabetic retina by modulating ASMase expression specifically in the retinal endothelial cells, we used three retinal cell types known to play a role in diabetes-induced pathology: human retinal endothelial cells (HREC), adult human retinal pigment epithelial cells (HRPE) and human Muller cells (HMC). We established that ASMase has the highest activity and protein level in HREC, when compared to HRPE and HMC. Immunohistochemistry assessment of ASMase distribution demonstrated that, although ASMase was ubiquitously expressed in the perinuclear intracellular compartment of all three cell types, HREC was the only cell type with pronounced plasma membrane ASMase expression. Moreover, DHA decreased ASMase activity only in HREC, when compared to HRPE and HMC. Importantly, DHA displaced ASMase from the caveolae membrane 119 microdomains in HREC and inhibited pro-inflammatory cytokine IL-IB induced lCAM-l expression. ASMase gene silencing in HREC recapitulated DHA effects on cytokine- induced endothelial cell activation. The effect of DHA on membrane microdomain sphingolipid composition in HREC was analyzed by tandem mass spectrometry. In support of ASMase inhibition and displacement of ASMase from caveolae membrane microdomains by DHA, we found a significant decrease in ceramide/sphingomyelin ratio in the caveolae fraction isolated from HREC treated with DHA. We have previously demonstrated that DHA prevents TNFa-induced ASMase activation and now we show that DHA prevents the TNFa-induced increase in the ceramide/sphingomyelin ratio in caveolar membrane microdomains. Taken together, these in vitro data provide a mechanism for the anti-inflammatory effect of DHA in endothelial cells, which we show to occur through regulation of caveolar ASMase expression. 120 2. Introduction Diabetic retinopathy is recognized as a microvascular complication of diabetes and there is evidence that diabetes compromises the survival of vascular cells (1). Very early stage diabetic retinopathy is regarded as a low-grade chronic inflammatory disease with leukocyte adhesion to the retinal vasculature, a process mediated by adhesion molecules expressed on the endothelial cell surface, especially intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-l) (2-4). Proinflammatory cytokines, including TNFa and IL-lB are increased in diabetic eyes (5- 7) and induce upregulation of adhesion molecule expression in vascular cells (8). Retinal endothelial cells become activated and then degenerate as diabetic retinopathy progresses (9, 10). Vascular endothelial cells are not the only cell type affected by diabetes in the retina. Muller cells, the main glial cells of the retina, are also dysfunctional in diabetes. Muller cells, analogous to brain astrocytes, synthesize factors that participate in formation of tight junction (11) and maintain the proper functioning of the inner blood-retinal barrier under normal conditions (11, 12); they also synthesize and store various grth factors with trophic and regulatory functions for a variety of cell types in the retina (13). Mizutani et al. provided evidence for selective biosynthetic modifications of Muller cells isolated from diabetic human eyes (1 3). Another retinal cell type affected by diabetes are the retinal pigment epithelial (RPE) cells that form the outer blood—retinal barrier. The RPE separates the neural retina from the fenestrated choriocapillaries and have a major role in controlling the flow of 121 solutes, nutrients and fluid from the choroidal vasculature to the outer retina (14, 15). Diabetes induces important morphological and biochemical changes in RPE cells (16-18). HREC contain lipid rafts and particular plasma membrane microdomains known as caveolae (19, 20) that have important roles in various physiological processes, especially in regulating vascular permeability (21) and signal transduction (22, 23). Caveolae are stabilized by the caveolins (24-27) and caveolin-1 is the primary structural protein in endothelial cells caveolae (28). Caveolae/lipid rafts microdomains are active assemblies of cholesterol, sphingolipids (sphingomyelin, ceramide) and glycerophospholipids (29). Sphingolipids are active components of membrane microdomains structure and are recognized to act as messengers in signaling pathways mediating inflammation, apoptosis, cell differentiation and proliferation (30). Sphingomyelinases are important early responders in inflammatory cytokine signaling that catalyze the hydrolysis of sphingomyelin to the bioactive second messenger ceramide (31-33). Acid sphingomyelinase (ASMase) is considered a major candidate for mediating stress-induced production of ceramide. ASMase is best known as a lysosomal enzyme where it plays an important role in lipid storage and sphingomyelin metabolism. Recent studies, however, identified an important role of ASMase in ceramide-mediated signal transduction. This unexpected function involves the translocation of ASMase from intracellular compartments to the outer leaflet of cell membranes. Hydrolysis of sphingomyelin into ceramide initiates a reorganization of the membrane which facilitates formation and coalescence of lipid microdomains (34, 35). Specifically, ASMase activity was localized to caveolae membrane microdomains (3 3). 122 Caveolae are enriched in sphingomyelin (26, 36-39) and thus represent a target for sphingomyelinase translocation and an important pool for ceramide production. Ceramide generation within the outer leaflet of the plasma membrane facilitates receptor clustering and enhances inflammatory signal transduction by several receptors that mediate the pathological changes typically associated with diabetic retinopathy, such as IL-lB and TNFa (40, 41). In this study, we examined a potential anti-inflammatory mechanism of DHA in HREC through differential regulation of caveolar versus lysosomal ASMase localization and promotion of sphingomyelin-enriched membrane microdomain formation with important consequences on cellular inflammatory signaling. 123 3. Materials and Methods 3.1. Reagents and Antibodies DMEM and F12 culture medium, antibiotics, fetal bovine serum and trypsin were obtained from Invitrogen (Carlsbad, CA). Amplex Red Sphingomyelinase Assay Kit, NuPAGE Novex 10% Bis- Tris gels, (Carlsbad, CA). GW4869 was purchased from Calbiochem (San Diego, CA). Commonly used chemicals and reagents were purchased fi'om Sigma (St. Louis, MO). Caveolin-1 and LAMP] antibodies (BD Bioscience, San Jose, CA), ICAM-l antibody (Santa Cruz Biotechnology, Santa Cruz, CA) were used. ASMase antibody was a generous gift from Dr. Richard Kolesnick. TNFa and IL-IB were from R&Dsystems (Minneapolis, MN). ON-TARGET plus SMART pool SMPDl (ASMase) and ON-TARGET plus siCONTROL (non-targeting pool, against luciferase) were purchased from Dharmacon (Chicago, IL). For lipid extraction and mass spectrometry, all solvents used were HPLC grade. Methanol (MeOH), and water were purchased from J.T. Baker (Phillipsburg, NJ). Ammonium hydroxide (NH4OH) and chloroform (CHC13) were from EMD Chemicals (Gibbstown, NJ). Isopropanol was from Fisher Scientific (Pittsburgh, PA). Lipid standards were obtained from Avanti Polar Lipids (Alabaster, AL). 3.2. Cell Culture Primary cultures of HREC, HRPE and HMC were prepared from postmortem human tissue (National Disease Research Interchange, Philadelphia, PA) cultured as previously described (42). Passages 1 to 5 were used in the experiments. For experimental treatment, 124 cells were transferred to serum-free medium for 14 to 24 hours before stimulatory agents were added. Only early passage cells were used in the experiments. 3.3. Fatty Acid Treatment The method has been previously described in chapter 11. BSA was used as a carrier control and linoleic acid, a (n6 polyunsaturated fatty acid was used as a lipid control. 3.4. Sphingomyelinase Assay The method has been previously described in chapter 11. 3.5. Western Blot Analysis The method has been previously described in chapter II. 3.6. Gene Silencing of ASMase The method has been previously described in chapter II. 125 3.7. Isolation of Caveolae/Lipid Rafts Plasma Membrane Microdomains a. The detergent-based method of caveolae/lipid raft isolation from HREC was previously described in chapter 11. b. Non—detergent method of isolation of caveolae/lipid rafts. Caveolae/lipid raft-enriched membrane microdomains were prepared using a slightly modified non-detergent OptiPrep gradient ultracentrifugation protocol (43). Briefly, 5 x 106 HREC were washed twice with cold PBS, collected into the base buffer (20 mM Tris-HCl, pH 7.8, 250 mM sucrose) to which has been added 1 mM CaC12 and 1 mM MgC12, Cells were eluted by centrifugation for two minutes at 250 g and resuspended in 0.33 ml of base buffer containing 1 mM CaC12 and 1 mM MgC12 and protease inhibitors. The cells were then lysed by passage through a 22gx3” needle 20 times. Lysates were centrifuged at 1,000g for 10 minutes. The resulting postnuclear supernatant was collected and transferred to a separate tube. The pellet was again lysed by the addition of 0.33 ml base buffer with divalent cations and protease inhibitors, followed by sheering 20 times through a 22gx3” needle. After centrifugation at 1,000g for 10 minutes, the second postnuclear supernatant was combined with the first. An equal volume (0.66 ml) of base buffer containing 50 % OptiPrep was added to the combined postnuclear supematants and placed on the bottom of a 4 m1 centrifuge tube. A 2.4 ml gradient of 0 % to 20 % OptiPrep in base buffer was poured on top of the lysate. Gradients were centrifuged for 18 hours at 200,000g at 4 °C using a swinging bucket rotor and 0.4 ml samples were collected carefully from the top of each fraction. A band confined to fractions one and two was designed as caveolae/lipid raft- enriched membrane domains. 126 3.8. Isolation of Plasma Membrane Domains The method has been previously described in chapter 11. 3.9. Lipid Extraction Lipid extraction from whole HREC. 3 x 106 HREC were washed twice with cold PBS, collected in 500 uL cold 40% methanol and homogenized in a glass homogenizer on ice. The cell homogenate was transferred to a screw-cap glass tube and lipids were extracted as previously described (44). The samples were dried under a stream of nitrogen and re- extracted twice to remove non-lipid contaminants. The lipid film was dried under nitrogen, then further dried overnight in a speedvac, and resuspended in an appropriate amount of isopropanol : methanol : chloroform (4 : 2 : 1) based on the protein concentration of each sample. Lipid extraction from HREC caveolae/lipid raft fractions and plasma membranes. For determination of sphingomyelin and ceramide content, plasma membranes and pooled caveolin-l enriched OptiPrep fractions were subjected to lipid extraction with alkaline hydrolysis of glycerophospholipids (45). Lipids were dried under a stream of nitrogen, and re-extracted twice as outlined above for whole HREC lipid extracts to remove non- lipid contaminants. Lipids were dried and resuspended as described above for whole HREC lipids. 127 3.10. Mass Spectrometry Analysis Synthetic lipid standards (Sphingolipid Mix 1, Avanti) were added to lipid extracts used in mass spectrometry analysis at 20 nmol/mg protein and 5 nmol/mg protein, respectively. Prior to analysis, lipids were appropriately diluted in isopropanol/methanol/ chloroform (4 : 2 : 1 v /v/ v) containing 20 mM ammonium hydroxide, centrifiiged, then loaded into Whatrnan Multichem 96-well plates (Fisher Scientific, Pittsburgh, PA) and sealed with Teflon Ultra-Thin Sealing Tape (Analytical Sales and Services, Pompton Plains, NJ). Lipids were introduced to a Thermo model TSQ Quantum Ultra triple quadrupole mass spectrometer (San Jose, CA) as previously described (44). Ceramide (Cer) and sphingomyelin (SM) molecular species were monitored as their [M+H]+ ions by tandem mass spectrometry (MS/MS) precursor ion scanning for the formation of characteristic product ions at m/z 264.4 (Cer) and m/z 184 (SM) in positive ionization mode. All MS/MS spectra were acquired automatically for 5-10 minutes at a rate of 500 m/z sec'l by methods created using Xcalibur software (Thermo, San Jose, CA). Correction for '3 C isotope effects and quantitation of lipid molecular species against appropriate internal standards was performed using the Lipid Mass Spectrum Analysis (LIMSA) software (46) peak model fit algorithm in conjunction with an expanded user- defined database of hypothetical lipid compounds. Ceramide and sphingomyelin molecular species were quantitated ratiometrically by comparison with the peak area of their synthetic lipid internal standard. The LIMSA software peak model fit algorithm was used as previously described for peak finding and isotope correction to arrive at an "absolute" value for each identified lipid species. All identified ceramide and sphingomyelin molecular species were summed in order to quantify the total ceramide 128 and total sphingomyelin amount in the samples. The final result for each sample was presented as ceramide/sphingomyelin ratio. MS/MS collision energies, CID gas pressure, and Q1 and Q3 settings were optimized using synthetic lipid standards corresponding to the individual lipid classes of interest. 3.11. Immunofluorescence Assay Cells were cultured on cover slips. The next day, cells on cover slides were removed from the media and rinsed in warm leBS three times. Then cells were fixed and permeabilized in ice-cold methanol for 15 minutes at -20°C, washed with warm leBS three times before blocking in a 2% BSA in leBS solution for one hour at 37°C, and then incubated with primary anti-Caveolin—l, anti-ASMase and anti-LAMP] antibodies and in a corresponding secondary antibody: Alexa Fluor 488 for Caveolin-land LAMPl and Alexa Fluor 495 for ASMase. All the primary and secondary antibodies were used in 1:100 dilutions. The cells were washed three times (five minutes each) with warm leBS before mounting the cover slips on the slides. ASMase and Caveolin-1 colocalization was visualized with a confocal fluorescence microscope and measured with Olympus Flroiew 1000 colocalization software. For images shown in Figure 3.2 B cells were fixed in 2% paraformaldehyde in PBS and where indicated, cells were permeabilized by ten minutes incubation in 0.1% TritonX-IOO. 129 3.12. Statistical Analysis Data are expressed as the mean i SD. Factorial ANOVA with post hoc Tukey test (GraphPad PrismS, GraphPad Software, San Diego, CA) was used for comparing the data obtained from independent samples. T-test was used to compare data obtained from two independent measurements. Significance was established at P<0.05. 130 4. Results 4.1. ASMase Expression and Activity in Human Retinal Cells ASMase activity and expression level was assessed in three human retinal cell cultures: human retinal endothelial cells (HREC), human adult retinal pigment epithelial cells (HRPE) and human Muller cells (HMC). In these three retinal cell types, we found that HREC have the highest ASMase activity (Figure 3.1 A) and protein levels (Figure 3.1 B,C) when compared to HRPE and HMC. 4.2. ASMase Cellular Distribution in Human Retinal Cells ASMase is ubiquitously expressed in the lysosomal compartment of all cell types. Lysosomal ASMase plays an important role in sphingomyelin metabolism but is not associated with inflammatory signaling. Recently endothelial cells were shown to secrete copious amounts of ASMase in response to inflammatory cytokines and the enzymatically active ASMase isofonn was found in the caveolae membrane microdomain compartment of these endothelial cells (33). Caveolae are highly enriched in sphingomyelin and 70% of total cellular sphingomyelin is found in these membrane microdomains (26, 36-39). In order to hydrolyze plasma membrane sphingomyelin to pro-inflammatory and pro-apoptotic ceramide, ASMase has to be present in the caveolae. Therefore, we investigated the cellular distribution of ASMase in HREC, HRPE and HMC. In HREC, we found that ASMase and caveolin-1, a marker for caveolae microdomains, colocalize both at the membrane level and within the cytoplasm 56.38i5.85 % of ASMase staining colocalizes with caveolin-1 (Figure 3.2 A, upper 131 panel); however, we also observed a strong punctuate perinuclear staining negative for caveolin-1, which likely represents lysosomal ASMase. To confirm that perinuclear ASMase lies within the lysosomal compartment, lysosomal associated membrane protein 1 (LAMPl) was used as a marker for this cellular compartment. We found that perinuclear ASMase closely associates with LAMPl (Figure 3.2 A, lower panel) as indicated by the yellow color in the merged panel. Moreover, although ASMase was ubiquitously expressed in the perinuclear intracellular compartment in HREC, HRPE and HMC (Figure 3.2 B,D), HREC were the only cell type with pronounced plasma membrane expression of ASMase (Figure 3.2 B,C) 4.3. DHA Effect on ASMase Activity in Human Retinal Cells As we have previously shown, DHA-pretreatment decreases basal ASMase activity in HREC. To compare the effect of DHA on ASMase activity on various retinal cell types, we next determined ASMase activity in HRPE and HMC that were grown in control media or media supplemented with DHA. DHA had no effect on ASMase activity in HRPE (Figure 3.3 A) and HMC (Figure 3.3 B) when compared to control cells. 4.4. Downregulation of caveolar ASMase by DHA Next we assessed ASMase colocalization with caveolin-1 in the caveolae/lipid raft fi'actions isolated from HREC by sucrose discontinuous gradient ultracentrifugation. Partial caveolae localization of ASMase was confirmed by western blot (Figure 3.4 A,C). 132 Moreover, DHA pretreatment of HREC prior to isolation of plasma membrane microdomains shows downregulation of ASMase in caveolae microdomains (Figure 3.4 B,C) with no effect on the ASMase expression in the intracellular compartment. 4.5. Sphingomyelin Enrichment in Caveolae/Lipid Rafts by DHA To address the effect of DHA on sphingolipid composition of caveolae/lipid rafts, these microdomains were isolated by OptiPrep continuous gradient ultracentrifugation then subjected to lipid extraction and lipid analysis by tandem mass spectrometry for sphingomyelin and ceramide molecular species. Continuation of isolated lipid rafts was achieved by showing enrichment of Caveolin-1 in density gradient fractions one and two. We found a significant decrease in ceramide/sphingomyelin ratio in the caveolae/lipid raft fi'actions isolated from HREC treated with DHA compared to BSA (vehicle control) or linoleic acid (lipid control) (Figure 3.5A). Moreover, DHA pretreated HREC prevented TNFa-induced increase in ceramide/sphingomyelin ratio (Figure 3.5 C). We found no significant modification of ceramide/sphingomyelin ratio in the plasma membrane domains isolated from control and DHA treated HREC (Figure 3.5 B). These data show that DHA-induced ASMase downregulation in the caveolae compartment results in formation of sphingomyelin-enriched caveolae/lipid rafts membrane microdomains. 133 4.6. Gene Silencing of ASMase Prevents Proinflammatory Cytokine-Induced HREC Activation Treatment of HREC with SngmL IL-IB for 6 hours induced dramatic upregulation of ICAM-1 protein expression (Figure 3.6 A,B). DHA significantly reduced cytokine- induced ICAM-l protein expression in HREC (Figure 3.6 A,B). More importantly, ASMase gene silencing recapitulated the effects of DHA on cytokine-induced adhesion molecules expression in HREC, preventing the IL-lB-induced increase in ICAM-l protein level in HREC (Figure 3.6 CD). 134 ASMase activtiy 1 .5 0.8 a) E5 a 0.6 r on E 5 0.4 ~ E . 0.2 -‘ 0 -1 --_ _. ___ , HREC HRPE HMC B C. .E , 5 f 8 0.2 1 t— f E ; HREC HRPE HMC 8 Q1 * * r _, v . \r .\ E, ; ASMase.fl.“;§.'-:;..a; g, 5, < L Tubulin mm 0 ,. . . _ HREC HRPE HMC Figure 3.1. ASMase expression in human retinal cells. Three human retinal cell cultures are shown: human retinal endothelial cells (HREC), human retinal pigment epithelial (HRPE) and human Muller (HMC) cells. (A) ASMase activity in HREC, HRPE and HMC. (B) ASMase expression in HREC, HRPE and HMC was determined by immunoblot. (C) Quantitative analysis of the data in panel (B) for ASMase normalized to tubulin level. The results are meaniSD of at least 3 independent experiments; *P<0.05 compared to HREC. 13S Human retinal endothelial cells Caveolin-1 l l ASMase l l Merged i Nucleus Nucleus a ar / ; Pefinucl V l Membrane f i Membrane . Perinuclear f Perinuclear . Membrane 7 Membrane Figure 3.2. ASMase cellular distribution in human retinal cells. Three human retinal cell cultures are shown: human retinal endothelial cells (HREC), human retinal pigment epithelial (HRPE) and human Muller (HMC) cells. (A) ASMase, Caveolin-1 and LAMPl colocalization in HREC was assessed by immunohistochemistry. There was 56.38i5.85 % colocalization of ASMase and caveolin-1 both at the plasma membrane level and in the cytoplasm (upper panel). However, there was strong punctuate perinuclear staining, negative for caveolin-1 that associates with LAMP] (lower panel), a marker for the lysosomal compartment (yellow color in the Merged panel). 136 Non-permeabilized Permeabilized ‘____INucleus O UJ 0: / I \ «Membrane NUCleUS ii." \ Nucleus CK I O 2 I / Nucleus Figure 3.2 (continued). ASMase cellular distribution in human retinal cells. (B) Immunofluorescent assessment of ASMase cellular distribution in non-penneabilized (left) and permeabilized (right) HREC, HRPE and HMC. 137 ASMase staining in non-permeabilized cells 'U S E E 20 8 cu g (D as g 10 ‘0 0 .~.-. 9:3... 1:1 nd nd 5 s o - r _S_.’ HREC HRPE HMC Q D. ASMase staining in permeabilized cells '0 .8 E“ .15 2° 8 g S as 5 10 ~ 2.9 E- I g .9 e .E g 0 -. f g HREC HRPE HMC Figure 3.2 (continued). ASMase cellular distribution in human retinal cells. (C) Quantification of ASMase staining in non-permeabilized cells corresponding to plasma membrane localization of ASMase in HREC, HRPE and HMC (40 fields per experiment). (D) Quantification of ASMase staining in permeabilized cells corresponding to ASMase in the lysosomal compartment in HREC, HRPE and HMC (40 fields per experiment). The results are meanISD of at least 3 independent experiments; nd= non- deterrnined (red calibration bar = 27.5um, blue calibration bar = 55pm, white calibration bar = llOum). 138 HRPE 300 100 ASMase activity/ ug protein (Arbitrary units) 0 3111_ Control HMC 150 100 “j 50, ASMase activity/ pg protein (Arbitrary units) 1--_,_-4_ 11.1.1. can: DHA Figure 3.3. The effect of DHA on ASMase activity in HRPE and HMC. ASMase activity in (A) HRPE and (B) HMC treated for 18 hours with 100 uM DHA compared to control. The results are meaniSD of three independent experiments. 139 A. Control Ca veolin-1 ASMase B- DHA treatment 1 Caveolin-1 ASMase ASMase protein level (Arbitrary units) Figure 3.4. Downregulation of caveolar ASMase by DHA. (A-C) ASMase protein expression level in caveolae fractions (fractions 3-4) versus non-caveolae fractions (fractions 5-9) in control (A and C, white bar) and DHA (B and D, gray bar)-treated HREC. The results are meaniSD of three independent experiments, +P<0.01 compared to control. Caveolae (fractions 3-4) (fractions 5-9) Fractions 2 3 4 5 6 7 8 9 - W ...-.. Fractions 2 3 4 5 6 7 8 9 "‘- “ 1...... “.1 .‘ ‘. . _. ASMase Non-caveolae 140 Ceramide/sphingomyelin ratio Membrane microdomains 0.08 .2 g 0.06 l 2* g 0.04 ~ ".5 I E 0.02 I Control DHA B. Ceramide/sphingomyelin ratio Plasma membrane ,9 0.08 ' s 0.06 ~ I 2‘ g 0.04 L5 2 0.02 -‘ Control DHA Figure 3.5. The effect of DHA on sphingomyelin and ceramide content of caveolae membrane microdomain in HREC. (A) Decrease in ceramide/sphingomyelin ratio in caveolae from DHA treated HREC compared to control. (B) No change in ceramide/sphingomyelin ratio in total plasma membrane. 141 Ceramide/sphingomyelin ratio Membrane microdomains i 0.1 ~ 0.05 % Arbitrary units cannot TNFa DHA+TNFa G Control HREC - membrane microdomains 12:0 Cer (l.S.) Ceramide .3 0 24:1 Cer 24:0 Cer 16:0 C6,,H20 1630 Cer 12:0 Cerebroside (15.) 25:0 Cer US.) l . l 480 500 520 540 560 580/ 600 620 640 660 680 700 mz Relative abundance (%) (II o 1 6:0 SM Sphingomyelin 24:1 SM 2108M 26:1 SM 24:2 SM _ 0 640 660 680 700 720 740 760 780 800 820 840 mlz Relative abundance (%) Figure 3.5 (continued). The effect of DHA on sphingomyelin and ceramide content of caveolae membrane microdomain in HREC. (C) DHA prevents TNFa-induced increase in ceramide/sphingo-myelin ratio in the caveolae. Tandem mass spectrum of control HREC (D), DHA treated HREC (E), TNFa treated HREC (F) and DHA and TNFa treated HREC (G) caveolae sphingomyelin species; and control HREC (H) and DHA treated HREC (I) of plasma membrane sphingomyelin species by positive ion mode PI m/z 184 after alkaline hydrolysis of glycerophospholipds. 142 I” DHA treated HREC - membrane microdomains g Ceramide a 12:0 Cer (1.8.) L5? 100 24:1 Cer g 24:0 Cer ‘9 50 . 25:0 Cer (I.S.) “ . 12:0 Cerebrosrde LS. . g 16:0 Cer-H2016-O Cer ( ) : *5 .1 6 l l . .l . . 0! 480 500 520 540 560 582/ 600 620 640 660 680 700 2 2\: 15:0 SM Sphingomyelin 8 100} C - g _ g 50; 24:1 SM 3 '120 SM (1 s ) 242 SM '240 SM 3 ' ' '1611 SM 18:0 SM ' 26:1 SM 59 . g 640 660 680 700 720 74/0 760 780 800 820 840 m 2 F“ TNFa treated HREC - membrane microdomains é Ceramide 0: 12:0 Cer (IS) 3 10' 24:1 Cer 53 24:0 Cer C {:3 5° 163° Cer 12:0 Cerebroside (I.S.) 253° 09' “-3) .f’>_’ 16'0 cepHZO ' 223° Cer 26:0 Cer E 1 . .l.. .. carJ 480 500 520 540 560 582/2 600 620 640 660 680 700 $3 :1; 10 16:0 SM Sphingomyelin C 8 24:1 SM .5. 0 3‘; 12:0 sM (1.5.) ; ' 242 SM 24:0 SM .3 l 16:1 SM < , 180 SM L 26:1 SM % _ . - l 1 .4*‘ f." I‘M Al -1 1 gullmll .1 1.5L. m 640 660 680 700 720 740 760 780 800 820 840 mlz Figure 3.5 (continued). The effect of DHA on sphingomyelin and ceramide content of caveolae membrane microdomain in HREC. 143 G. DHA and TNFa treated HREC - membrane microdomains 3 . 2, . Ceramide 8100 12.0Cer(l.8.) 2431C” g 24:0 Cer 5 25:0 Cer (1.8.) 3 50 - 8 16-0 09' 12:0 Cerebroside (1.8.) .QZ’ 16:0 Cer-H20 ‘ ' E C?!) 480 500 520 540 560 580 600 620 640 660 680 700 m/z 015 S h'ngo el' V . p I my In §100 16.0 SM (U "U C .8 <0 50 a) 12:0 SM 1.8. sf; | ( )16z18MHl l16:03M l l 24:1 SM '0') 05 640 660 680 700 720 740 760 780 800 820 840 mlz H' Control HREC - plasma membrane 24‘ V - Ceramide g 100 12.0 Cer (1.8.) 24:1 Cer "8“ 24:0 Cer g 50 16 0 c 25:0 Cer (1.8.) I er ' ' g 16:0 Cer-H20 12.0 Cerebrosrde (1.8.) E (“f 480 500 520 540 560 580/ 600 620 640 660 680 700 m Z Ix; V 16:0 8M ' ' 8 1 003 Sphingomyelin g - g 50: 12'08M(18 ) 24:1SM ‘0 l ' ‘ ' 24:2 8M 3 14:0 SM 1618M l 17:0 SM 24:0 SM .2 ‘: I - 22:0 8M 5 E . L. L 1 4h. .11., prSM . flu. in- . #1 11 6:9 640 660 680 700 720 740 760 780 800 820 840 mlz Figure 3.5 (continued). The effect of DHA on sphingomyelin and ceramide content of caveolae membrane microdomain in HREC. 144 [_DHA treated HREC - plasma membrane § V . Ceramide g 100 12.0Cer(l.8.) 24:1Cer 'c ‘ 24ZOCer C , _. é 12:0Cerebroside(l.8.) 5 .’ 25:00er(|-S.) 0) 16:0 Cer 1‘ ,.>_., 16:0 Cer-H20 s 0) I “I : a - ‘: ‘1 480 500 520 540 560 58,2, 600 620 640 660 680 700 Z :5 E 100 16:0 8M Sphingomyelin g 50 2418M 53 1208M (IS) 17:08M 24325” > . . . '8 L140s~1161 SM 18:0 SM 22:0 SM “.1340“ SM 6 a. - _ . 4.. . . - ‘1 640 660 680 700 720 74,0 760 780 800 820 840 "'12 Figure 3.5 (continued). The effect of DHA on sphingomyelin and ceramide content of caveolae membrane microdomain in HREC. Tandem mass spectrum of control HREC (D), DHA treated HREC (E), TNFa treated HREC (F) and DHA and TNFa treated HREC (G) caveolae ceramide species; and control HREC (H) and DHA treated HREC (I) of plasma membrane ceramide species by positive ion mode PI m/z 264. The results are meaniSD of three independent experiments performed in triplicates, *P<0.05 compared to control, #P<0.05 compared to control and DHA treated and TNFa stimulated HREC. 145 BSA + + + DHA - - + lL-1B - + + lCAM-l d an... Tubulin — - -1 B: ICAM-1 A O ,1. .1_._I__1_.___l__ .. .. 5 30 a is” / 8 ,9 // .E 'o 20 , ‘ a E 10 / g A 0 .____'I.'_ 244‘ v. 08 lL-1B Figure 3.6. The effect of DHA and ASMase gene silencing on IL-lB-induced HREC activation. (A) DHA or (B) ASMase siRNA treatment prevents IL-lB induced increase in [CAM-1 expression in HREC. Random siRNA was used as control for ASM siRNA experiments. The results are meaniSD of at least three independent experiments performed in triplicates, *P<0.05 compared to control; #P<0.05 compared to IL-18 stimulated HREC, IP<0.05 compared to control. 146 Electroporation + + + + lL-1B - + + + siRNA control - - + - siRNA ASMase - - - + lCAM-‘l _ -— *- Tubulin --- D. ICAM-1 60 g T * * .E 1 2 ' , g. E 3 40 fl}: “ 9 .9 fit/3 ' .1 g ”g a; --' .5 i; E To 20 “ f/fi 2; E /// = 8 = K 0 K: E \ 0 I I $0 09 0 Figure 3.6 (continued). The effect of DHA and ASMase gene silencing on IL-lll- induced HREC activation. 147 5. Discussion The fluid mosaic model of the cell membrane structure proposed by Singer and Nicholson in 1972 implied that membranes are in a disordered state with no major selectivity (47). However, recently , an extensive literature has showed the existance of microdomains that exist in a liquid-ordered phase within the membrane structure (48). These membrane microdomains are highly enriched in sphingolipids (sphingomyelin, ceramides) and cholesterol, are static within the membrane, but have the ability to coalesce and rearrange in response to diverse stimuli (49). The generation of ceramide at the plasma membrane level dramatically alters the properties of the cell membrane, since ceramide molecules have the tendency to spontaneously self-associate (50, 51). It is generally accepted that ceramide-rich membrane microdomains could primarily function to reorganize receptors and various signaling molecules at the plasma membrane level in order to facilitate and amplify cellular signaling processes via specific receptors (52). Sphingomyelinases are sphingolipid hydrolyses that catalyze the breakdown of sphingomyelin to ceramide and phosphorylcholine. Several isoforms of sphingomyelinases have been identified and further distinguished by their catalytic pH optimum, cellular localization, primary structure and co-factor dependence. Alkaline sphingomyelinase activity is confined to the intestinal mucosa, bile and liver and does not participate in signal transduction (36, 53, 54). Neutral sphingomyelinase (NSMase) and ASMase are crucially involved in the pathophysiology of metabolic disorders (55) and play an active role in cellular signaling (56). ASMase is classically known to localize to the lysosomal compartment. Lysosomal ASMase is ubiquitously expressed in all cell types. However, several studies 148 also identified a plasma membrane-associated form of ASMase in human fibroblasts (33) and pulmonary vascular endothelial cells (57). Another study brought out evidence of the existence of a small pool of zinc-independent ASMase in the plasma membrane microdomains purified from NIH-3T3 L1 fibroblasts and PC12 cells and more importantly, this pool of ASMase could undergo activation in response to neurotrophins, whereas the lysosomal ASMase did not respond to activation by ligand (58). Various immunohistochemistry studies have shown that cells exposed to different stress stimuli exhibit a change in ASMase location from intracellular compartment to the cell surface (59). Plasma membrane-associated ASMase, rather than intracellular ASMase, is shown to be associated with inflammatory signaling (49), as ceramide generation within the plasma membrane results in coalescence and reorganization of membrane microdomains into large signaling platforms that facilitate receptor clustering and cellular signal transduction (60, 61). Endothelial cells are well recognized to be an important source of ASMase, expressing as much as 20 times more ASMase as other mammalian cell types (62, 63). Human coronary artery and human umbilical vein endothelial cells, as well as bovine and murine aortic endothelial cells secrete copious amounts of ASMase in response to inflammatory cytokines (63). Our study also shows that HREC exhibit high ASMase activity and expression level when compared to other retinal tissues, including HRPE and HMC cells (Figure 3.1 A,B,C). Moreover, in this study we demonstrate that it is this plasma membrane ASMase that is a key element in the retinal inflammatory response in HREC; HREC was the only cell type to demonstrate positive staining for the plasma membrane form of ASMase, whereas HRPE and HMC presented only the classical, 149 intracellular form of ASMase. We have found in our study that 56.3 8i5.85 % of ASMase colocalize with caveolin-1 both at the membrane and intracellular levels and 44 % may represent the lysosomal form of ASMase. In endothelial cells, localization of ASMase to the plasma membrane, and particularly to caveolae microdomains, increases substrate availability for this enzyme. Omega 3 PUFAS, and especially DHA, have been long accepted to modulate inflammatory processes (64). We have previously shown that DHA pretreatment of HREC decrease ASMase activity and gene expression (65). Our present study demonstrates that DHA downregulates the ASMase protein level in the caveolae microdomain compartment, with no effect on intracellular ASMase protein level. Decreased ASMase expression in caveolae would profoundly impact the ability of HREC to generate ceramide in this membrane microdomain. Although DHA has disparate effects on the caveolar and lysosomal compartments, this differential sensitivity to DHA is likely due to a higher exchange and/or degradation rate of the caveolar ASM, which is dynamically regulated, compared to lysosomal ASM, which is constitutively expressed. Indeed, regulation of caveolar ASMase expression has a profound impact on membrane microdomains lipid composition, particularly on sphingomyelin and ceramide content. As we have demonstrated in this study, downregulation of caveolar ASMase by DHA induced a significant reduction in ceramide/sphingomyelin ratio and decreased the ability of the proinflammatory cytokine TNFa to induce ceramide production in these specialized membrane microdomains. However, as shown in the previous chapter, DHA also downregulates the activity and cellular expression of NSMase, another critical enzyme capable of sphingomyelin hydrolysis at the membrane level. Therefore, the DHA 150 effect on microdomain sphingolipid composition may be due to downregulation of both ASMase and NSMase. Yet, inhibition and/or gene silencing of ASMase rather than NSMase had a more pronounced effect on decreasing cytokine-induced adhesion molecules expression in HREC. In addition, several studies have downplayed the role of NSMase in ceramide-enriched platforms formation and transmembrane signal transmission (66, 67). Taken together, our results suggest that ASMase could play a more important role in cytokine-induced formation of ceramide-rich membrane microdomains and amplification of inflammatory signaling in our cell culture system. In conclusion, we demonstrated that HREC have the highest cellular ASMase expression and protein level when compared to HRPE and HMC. Moreover, the plasma membrane ASMase was expressed only in HREC. DHA downregulated caveolar ASMase expression and promoted formation of sphingomyelin-enriched membrane microdomains. These modified caveolae/lipid rafts have a decreased response to cytokines known to be upregulated in diabetic retina (TNF-a and IL-IB) as confirmed by decreased cellular adhesion molecules expression in HREC. Based on the results obtained in these in vitro studies, we addressed in the following chapter the role of sphingomyelinases in retinal vascular inflammation and vessel loss in diabetic retinopathy, using a streptozotocin (STZ)-induced diabetes rat model; we also characterized the effect of DHA supplementation on retinal sphingomyelinase expression, inflammatory status and microangiopathy lesions in this type 1 diabetic rat model. 151 6. References 10. Mizutani, M., Kern, TS, and Lorenzi, M. 1996. Accelerated death of retinal microvascular cells in human and experimental diabetic retinopathy. J Clin Invest 97:2883-2890. McLeod, D.S., Lefer, D.J., Merges, C., and Lutty, GA. 1995. Enhanced expression of intracellular adhesion molecule-1 and P-selectin in the diabetic human retina and choroid. Am J Pathol 147:642-653. Joussen, A.M., Murata, T., Tsujikawa, A., Kirchhof, B., Bursell, S.E., and Adamis, AP. 200]. Leukocyte-mediated endothelial cell injury and death in the diabetic retina. Am J Pathol 158:147-152. Adamis, AP. 2002. Is diabetic retinopathy an inflammatory disease? Br J Ophthalmol 86:363-365. Limb, G.A., Chignell, A.H., Green, W., LeRoy, F., and Dumonde, DC. 1996. Distribution of TNF alpha and its reactive vascular adhesion molecules in fibrovascular membranes of proliferative diabetic retinopathy. Br J Ophthalmol 80:168-173. Joussen, A.M., Poulaki, V., Le, M.L., Koizumi, K., Esser, C., Janicki, H., Schraermeyer, U., Kociok, N., Fauser, S., Kirchhof, B., et al. 2004. A central role for inflammation in the pathogenesis of diabetic retinopathy. Faseb J 18:1450— 1452. Adamis, A.P., Miller, J .W., Bemal, M.T., D'Amico, D.J., Folkman, J ., Yeo, T.K., and Yeo, K.T. 1994. Increased vascular endothelial growth factor levels in the vitreous of eyes with proliferative diabetic retinopathy. Am J Ophthalmol 118:445-450. Chen, W., Esselman, W.J., Jump, DB, and Busik, J .V. 2005. Anti-inflammatory effect of docosahexaenoic acid on cytokine-induced adhesion molecule expression in human retinal vascular endothelial cells. Invest Ophthalmol Vis Sci 46:4342-4347. Miyamoto, K., Khosrof, S., Bursell, S.E., Rohan, R., Murata, T., Clermont, A.C., Aiello, L.P., Ogura, Y., and Adamis, AP. 1999. Prevention of leukostasis and vascular leakage in streptozotocin-induced diabetic retinopathy via intercellular adhesion molecule-1 inhibition. Proc Natl Acad Sci U S A 96: 10836-10841. Nozaki, M., Ogura, Y., Hirabayashi, Y., Saishin, Y., and Shimada, S. 2002. Enhanced expression of adhesion molecules of the retinal vascular endothelium in spontaneous diabetic rats. Ophthalmic Res 34: 1 58-164. 152 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. Tout, S., Chan-Ling, T., Hollander, H., and Stone, 1. 1993. The role of Muller cells in the formation of the blood-retinal barrier. Neuroscience 55:291-301. Distler, C., and Dreher, Z. 1996. Glia cells of the monkey retina-—II. Muller cells. Vision Res 36:2381-2394. Mizutani, M., Gerhardinger, C., and Lorenzi, M. 1998. Muller cell changes in human diabetic retinopathy. Diabetes 47:445-449. Erickson, K.K., Sundstrom, J .M., and Antonetti, D.A. 2007. Vascular permeability in ocular disease and the role of tight junctions. Angiogenesis 10:103-117. Strauss, O. 2005. The retinal pigment epithelium in visual function. Physiol Rev 85:845-881. Caldwell, R.B., and Slapnick, SM. 1989. Increased cytochrome oxidase activity in the diabetic rat retinal pigment epithelium. Invest Ophthalmol Vis Sci 30:591- 599. Grimes, P.A., and Laties, AM. 1980. Early morphological alteration of the pigment epithelium in streptozotocin-induced diabetes: increased surface area of the basal cell membrane. Exp Eye Res 30:631-639. Tso, M.C., Cunha-Vaz, J .G., Shih, C.Y., and Jones, C.W. 1980. Clinicopathologic study of blood-retinal barrier in experimental diabetes mellitus. Arch Ophthalmol 98:2032-2040. Mora, R.C., Bonilha, V.L., Shin, B.C., Hu, J., Cohen-Gould, L., Bok, D., and Rodriguez-Boulan, E. 2006. Bipolar assembly of caveolae in retinal pigment epithelium. Am J Physiol Cell Physiol 290:C832-843. Palade, GE. 1953. Fine structure of blood capillaries. J Appl Physiol 2421424— 1436. Miyawaki-Shimizu, K., Predescu, D., Shimizu, J ., Broman, M., Predescu, S., and Malik, AB. 2006. siRNA-induced caveolin-1 knockdown in mice increases lung vascular permeability via the junctional pathway. Am J Physiol Lung Cell Mol Physiol 2902L405-413. Anderson, R.G. 1993. Caveolae: where incoming and outgoing messengers meet. Proc Natl Acad Sci U S A 90: 10909-10913. 153 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. Lisanti, M.P., Tang, Z., Scherer, P.B., Kubler, E., Koleske, A.J., and Sargiacomo, M. 1995. Caveolae, transmembrane signalling and cellular transformation. Mol Membr Biol 122121-124. Anderson, R.G., and Jacobson, K. 2002. A role for lipid shells in targeting proteins to caveolae, rafis, and other lipid domains. Science 296:1821-1825. Razani, B., and Lisanti, MP. 2001. Caveolins and caveolae: molecular and functional relationships. Exp Cell Res 271:36-44. Smart, B.J., Graf, G.A., McNiven, M.A., Sessa, W.C., Engelman, J.A., Scherer, P.B., Okamoto, T., and Lisanti, MP. 1999. Caveolins, liquid-ordered domains, and signal transduction. Mol Cell Biol 19:7289—7304. Sotgia, F., Razani, B., Bonuccelli, G., Schubert, W., Battista, M., Lee, H., Capozza, F., Schubert, A.L., Minetti, C., Buckley, J .T., et al. 2002. Intracellular retention of glycosylphosphatidyl inositol-linked proteins in caveolin-deficient cells. Mol Cell Biol 22:3905-3926. Glenney, J.R., Jr. 1989. Tyrosine phosphorylation of a 22-kDa protein is correlated with transformation by Rous sarcoma virus. J Biol Chem 264:20163- 20166. Edidin, M. 2003. The state of lipid rafts: from model membranes to cells. Annu Rev Biophys Biomol Struct 32:257-283. Mathias, S., Pena, L.A., and Kolesnick, RN. 1998. Signal transduction of stress via ceramide. Biochem J 335 ( Pt 3):465-480. Bilderback, T.R., Gazula, V.R., Lisanti, M.P., and Dobrowsky, RT. 1999. Caveolin interacts with Trk A and p75(NTR) and regulates neurotrophin signaling pathways. J Biol Chem 274:257-263. Cremesti, A., Paris, F ., Grassme, H., Holler, N., Tschopp, J ., Fuks, Z., Gulbins, E., and Kolesnick, R. 2001. Ceramide enables fas to cap and kill. J Biol Chem 276:23954-23 961 . Liu, P., and Anderson, R.G. 1995. Compartmentalized production of ceramide at the cell surface. J Biol Chem 270:27179-27185. Gulbins, E., and Kolesnick, R. 2003. Raft ceramide in molecular medicine. Oncogene 22:7070-7077. Gulbins, E., and Li, PL. 2006. Physiological and pathophysiological aspects of ceramide. Am J Physiol Regul Integr Comp Physiol 290:R1 1-26. 154 36. 37. 38. 39. 40. 41. 42. 43. 45. 46. 47. 48. Kolesnick, R. 2002. The therapeutic potential of modulating the ceramide/sphingomyelin pathway. J Clin Invest 110:3-8. Pierini, L.M., and Maxfield, F .R. 2001. Flotillas of lipid rafts fore and aft. Proc Natl Acad Sci U S A 98:9471-9473. Shaul, P.W., and Anderson, R.G. 1998. Role of plasmalemmal caveolae in signal transduction. Am J Physiol 27 5:L843-851 . Simons, K., and Ikonen, E. 1997. Functional rafts in cell membranes. Nature 387:569-572. Ko, Y.G., Lee, J.S., Kang, Y.S., Ahn, J.H., and Seo, J.S. 1999. TNF-alpha- mediated apoptosis is initiated in caveolae-like domains. J Immunol 162:7217- 7223. Martin, M.U., and Wesche, H. 2002. Summary and comparison of the signaling mechanisms of the Toll/interleukin-l receptor family. Biochim Biophys Acta 1592:265-280. Busik, J .V., Mohr, S., and Grant, MB. 2008. Hyperglycemia-induced reactive oxygen species toxicity to endothelial cells is dependent on paracrine mediators. Diabetes 57:1952-1965. Macdonald, J .L., and Pike, L]. 2005. A simplified method for the preparation of detergent-free lipid rafts. J Lipid Res 46:1061-1067. Lydic, T.A., Busik, J.V., Esselman, W.J., and Reid, GE. 2009. Complementary precursor ion and neutral loss scan mode tandem mass spectrometry for the analysis of g1ycerophosphatidylethanolamine lipids from whole rat retina. Anal Bioanal Chem. Merrill, A.H., Jr., Sullards, M.C., Allegood, J.C., Kelly, S., and Wang, E. 2005. Sphingolipidomics: hi gh-throughput, structure-specific, and quantitative analysis of sphingolipids by liquid chromatography tandem mass spectrometry. Methods 36:207-224. Haimi, P., Uphoff, A., Hermansson, M., and Somerharju, P. 2006. Software tools for analysis of mass spectrometric lipidome data. Anal Chem 78:8324-8331. Singer, S.J., and Nicolson, G.L. 1972. The fluid mosaic model of the structure of cell membranes. Science 1 75 :7 20-73 1 . Jacobson, K., Sheets, ED, and Simson, R. 1995. Revisiting the fluid mosaic model of membranes. Science 268:1441-1442. 155 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. Schuchman, EH. 2009. Acid sphingomyelinase, cell membranes and human disease: Lessons from Niemann-Pick disease. FEBS Lett. Holopainen, J .M., Subramanian, M., and Kinnunen, PK. 1998. Sphingomyelinase induces lipid microdomain formation in a fluid phosphatidylcholine/sphingomyelin membrane. Biochemistry 3 7:1 7562-17570. Kolesnick, R.N., Goni, F.M., and Alonso, A. 2000. Compartrnentalization of ceramide signaling: physical foundations and biological effects. J Cell Physiol 184:285-300. Grassme, H., Riethmuller, J., and Gulbins, E. 2007. Biological aspects of ceramide-enriched membrane domains. Prog Lipid Res 46:161-170. Duan, RD. 2006. Alkaline sphingomyelinase: an old enzyme with novel implications. Biochim Biophys Acta 1761:281-291. Pavoine, C., and Pecker, F. 2009. Sphingomyelinases: their regulation and roles in cardiovascular pathophysiology. Cardiovasc Res 82:175-183. Holland, W.L., and Summers, SA. 2008. Sphingolipids, insulin resistance, and metabolic disease: new insights from in vivo manipulation of sphingolipid metabolism. Endocr Rev 29:381-402. Marchesini, N., and Hannun, Y.A. 2004. Acid and neutral sphingomyelinases: roles and mechanisms of regulation. Biochem Cell Biol 82:27-44. Yang, Y., Yin, J ., Baumgartner, W., Samapati, R., Solymosi, E.A., Reppien, E., Kuebler, W.M., and Uhlig, S. 2009. Platelet-activating factor reduces endothelial NO production - role of acid sphingomyelinase. Eur Respir J. Dobrowsky, RT. 2000. Sphingolipid signalling domains floating on rafts or buried in caves? Cell Signal 12281-90. Gulbins, E. 2003. Regulation of death receptor signaling and apoptosis by ceramide. Pharmacol Res 47:393-399. Grassme, H., Jendrossek, V., Riehle, A., von Kurthy, G., Berger, J ., Schwarz, H., Weller, M., Kolesnick, R., and Gulbins, E. 2003. Host defense against Pseudomonas aeruginosa requires ceramide-rich membrane rafts. Nat Med 9:322- 330. Gulbins, E., and Grassme, H. 2002. Ceramide and cell death receptor clustering. Biochim Biophys Acta 1585:139-145. 156 62. 63. 64. 65. 66. 67. Andrieu-Abadie, N., and Levade, T. 2002. Sphingomyelin hydrolysis during apoptosis. Biochim Biophys Acta 1585: 1 26-134. Marathe, S., Schissel, S.L., Yellin, M.J., Beatini, N., Mintzer, R., Williams, K.J., and Tabas, I. 1998. Human vascular endothelial cells are a rich and regulatable source of secretory sphingomyelinase. Implications for early atherogenesis and ceramide-mediated cell signaling. J Biol Chem 273:4081-4088. Calder, P.C. 2006. n-3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr 83:1505S-1519S. Opreanu, M., Lydic, T.A., Reid, G.E., McSorley, K.M., Esselman, W.J., and Busik, J. 2010. Docosahexaenoic Acid Inhibits Cytokine Signaling in Human Retinal Endothelial Cells by Downregulating Sphingomyelinases. Invest Ophthalmol Vis Sci. Grassme, H., Jekle, A., Riehle, A., Schwarz, H., Berger, J., Sandhoff, K., Kolesnick, R., and Gulbins, E. 2001. CD95 signaling via ceramide-rich membrane rafts. J Biol Chem 276:20589-20596. Veldman, R.J., Maestre, N., Aduib, G.M., Medin, J .A., Salvayre, R., and Levade, T. 2001. A neutral sphingomyelinase resides in sphingolipid-enriched microdomains and is inhibited by the caveolin-scaffolding domain: potential implications in tumour necrosis factor signalling. Biochem J 355:859-868. 157 Chapter IV Docosahexaenoic Acid Supplementation Prevents Upregulation of Proinflammatory/Angiogenic Molecules and Vessel Loss in Diabetic Retina Through Downregulation of Acid Sphingomyelinase I . Abstract Sphingomyelinases (SMases) are key regulatory enzymes of sphingolipid metabolism, catalyzing hydrolysis of sphingomyelin to the proinflammatory and pro- apoptotic messenger ceramide. In this study, we addressed the role of SMases in retinal vascular inflammation and vessel loss in diabetic retinopathy, using a streptozotocin (STZ)-induced diabetes rat model. Acid (ASMase), but not neutral (N SMase) SMase was upregulated in the retinas with microangiopathy lesions. Proinflarnmatory/angiogenic molecules were significantly increased in retina of STZ rats at one month and nine month post STZ treatment. However, a docosahexaenoic acid (DHA)-enriched diet restored ASMase in diabetic retina to the control levels and prevented upregulation of inflammatory/angiogenic mediators’ gene expression, strongly demonstrating a DHA protective effect against the development of an early inflammatory status in the diabetic retina. More importantly, DHA prevented retinal vessel loss in the retina of STZ treated, diabetic rats. In order to confirm that the protective effect of DHA in the retina is mediated through vascular ASMase, we next used an ischemia/reperfusion (I/R) injury model of retinopathy in rats. Immunohistochemistry analysis of normal rat retina demonstrated that retinal vasculature preferentially express ASMase when compared the 158 other retinal tissues. Furthermore, rat retinal I/R specifically increased ASMase expression in the vasculature, with little or no effect on ASMase expression in other retinal cells. Notably, DHA supplementation prior to retinal injury prevented ASMase upregulation in the retinal vasculature. Taken together, these in vivo data demonstrated ASMase downregulation as a mechanism by which DHA exerts its protective effect against retinal microvascular damage in diabetic retinopathy. 159 2. Introduction Visual impairment among people with diabetic retinopathy is a major disability with a profound impact on quality of life (1). Diabetic retinopathy (DR) represents the ocular expression of diabetic microangiopathy. The natural history of DR has been classified into an early, non-proliferative stage (background retinopathy) and a later, proliferative stage. The early stage of DR has distinct vascular lesions that are histologically characterized by the existence of saccular capillary microaneurysms, retinal hemorrhage, venous dilation and beading, retinal lipid exudates, pericyte degeneration, occlusion and degeneration of retinal capillaries (2-3). This early stage diabetic retinopathy represents a persistent low-grade chronic inflammatory disease (4) that involves leukocyte recruitment and adhesion to the retinal vasculature and up-regulation of inflammatory genes. Adhesion molecules, especially intercellular adhesion molecule-1 (ICAM-l), are involved in leukocyte attachment and transmigration into the vascular intima (5-6) that will result in early blood retinal barrier breakdown, capillary non- perfusion, endothelial cell injury and death. Proinflammatory cytokines (TNFa, IL-IB, IL-6 and VEGF) are increased in diabetic eyes and several inflammatory pathways are activated in the very early stages of diabetic retinopathy (7-11). Proinflammatory cytokines that are upregulated in diabetic eyes, including TNFa and IL-lB have been shown to activate cellular sphingomyelinases in various cell types (12-15), including retinal vascular endothelial cells, as presented in the previous chapters. Sphingomyelinases are enzymes that catalyze the hydrolysis of sphingomyelin (ceramide phosphocholine) to the bioactive lipid ceramide. Caveolae microdomains present on endothelial cells are enriched in sphingomyelin and thus represent an important pool for 160 ceramide production. Ceramide generation within the outer leaflet of the plasma membrane facilitates receptor clustering and enhances inflammatory signal transduction by several receptors that mediate the pathological changes typically associated with diabetic retinopathy, such as IL-IB and TNFa (16-17). As previously underlined, vascular endothelial cells represent the target tissue affected by diabetic retinopathy and become dysfunctional and degenerate due to diabetic metabolic dysregulation and inflammatory stress. Moreover, vascular endothelial cells are considered to be a very rich source of secreted ASMase and pro-inflammatory cytokines, especially IL-l, increase ASMase secretion in in vivo studies (18). The mechanisms that induce the proinflammatory state in the retina are not well defined, but various studies have shown that hyperglycemia and dyslipidemia play an important role. Hyperglycemia, a major causative factor in the development of diabetic retinopathy, induces cytokine production by several distinct retinal cell types including retinal pigment epithelium, Muller cells, astrocytes, microglia and pericytes, but interestingly not by retinal endothelial cells. We previously demonstrated in vitro that retinal endothelial cells increase the production of reactive oxygen species in response to diabetes induced cytokine release by other retinal cells rather than directly to high glucose, suggesting that in vivo diabetes-related endothelial injury in the retina is also due to glucose-induced cytokine release and not a direct effect of high glucose on endothelial cells (19). Retinal endothelial cells express TNF-a and IL-IB receptors and stimulation with cytokines activate classic NF KB inflammatory pathways leading to increase expression of inflammatory genes, including adhesion molecules ICAM-1 and VCAM-l. 161 In addition to hyperglycemia, Diabetes Control and Complications and Epidemiology of Diabetes Interventions and Complications Trials (DCCT/EDIC) demonstrated the strong association between dyslipidemia and development of diabetic retinopathy. We have previously demonstrated that the retina has a unique fatty acid composition exhibiting the highest level of (03-polyunsaturated fatty acids (PUFA), especially DHA, in the body and that retinal fatty acid metabolism is distinct from the metabolic changes that occur in diabetic plasma or diabetic liver (20). Omega 3 PUFAS have the ability to modulate various biological processes involved in retinal vascular inflammation, retinal capillary structure and integrity, and retinal angiogenesis (21). We have previously shown that DHA suppresses cytokine induced inflammatory signaling and activation of HREC (22). This pronounced anti- inflammatory effect of DHA was mediated through a reduction in ASMase and NSMase activity and expression (23). Importantly, total m3-PUFAs, and particularly DHA, are reduced in the retina of human (24) and rat (20) diabetic eyes. Moreover, increasing the retinal levels of (o3-PUFAs by genetic or dietary means has a beneficial effect in reducing pathological retinal angiogenesis (21). Activation of sphingomyelinases is an important early step in inflammatory cytokine signaling (13-14) particularly in endothelial cells (15, 23). In this study, we tested the hypothesis that downregulation of SMases by DHA may be protective against endothelial activation, injury and death, and thereby preventing retinal vessel loss in a rat model of type 1 diabetes. 162 3. Materials and Methods 3.1. Reagents and Antibodies Crude trypsin was obtained from Difco (Detroit, MI)4.. Lyophilized pancreatic elastase was purchased from Calbiochem (San Diego, CA). Streptozotocin (Sigma, St. Louis, MO), Neutral Protamine Hagedom (NPH) insulin, experimental diets Dyets Inc. (Bethlehem, PA), RT-PCR primers from IDT DNA Technologies (Coralville, IA) (Table 4.4 and Table 4.5). ASMase antibody was a generous gift from Dr. Richard Kolesnick. VEGF antibody was purchased from Novus Biologicals (Littleton, CO). NuPAGE Novex 10% Bis- Tris gels, Platinum SYBR Green qPCR SuperMix-UDG w/ROX, were purchased from Invitrogen (Carlsbad, CA). lRDye infiared secondary antibodies were purchased from Invitrogen, Molecular probes (Eugene, OR). 3.2. Rat Model of Type 1 Diabetic Retinopathy Male Sprague-Dawley rats weighing 237-283 grams were made diabetic with a single intraperitoneal injection of 65 mg Streptozotocin (STZ) (Sigma, St. Louis, MO) per kg body weight. STZ is a pancreatic B-cell cytotoxin, similar to glucose, transported into the cell by the glucose transport protein GLUT2. Starting with day 7 after STZ injection, Neutral Protamine Hagedom (NPH) insulin injections (with a dose of 0-3 units/day) was administrated in order to achieve slow weight gain, but allowing hyperglycemia in the range of 20mM blood glucose. The weight gain and blood glucose measurements from all the groups studied are presented in Table 4.1 and Table 4.2. The retina was harvested and analyzed at several time points (one month and nine months of diabetes). 163 3.3. The Experimental Diet In our study, we maintained the recommended 10% caloric intake from lipid and replaced 5% of this value with fish oil in the treatment group. Two dietary experimental groups of animals were established in both control non-diabetic and diabetic animals: a fish oil diet (co-3-PUFA) and a vegetable oil diet group. The diets were purchased from Dyets Inc. (Bethlehem, PA) and are based on AIN-93M purified rodent diet composition with 10% caloric intake as soybean oil (standard rodent diet ingredient contains 50.8% linoleic acid) or 5% as soybean oil and 5% as Menhaden oil (oil from a small plankton eating fish containing 10.26% DHA and 14.16% EPA). The n6/n3 fatty acid ratio in vegetable oil diet is 8/1, reflecting 15-16.7/1 ratio found in most Western diets (25). The n6/n3 ratio in fish oil diet represents 1.5/1, reflecting the ratios associated with prevention of cardiovascular disease (26) or suppression of inflammation in patients with rheumatoid arthritis (27). The fatty acid composition of the control, standard diet and fish oil (DHA)- enriched diet are presented in Table 4.3. 3.4. Isolation of Retinal Vasculature Rat retinal vasculatures were isolated by trypsin digestion method as previously described (28-29). The eyes were fixed in formalin for two weeks. The day before trypsin digest, the eyes were opened coronally, cornea and lens were discarded and posterior globe was placed into distilled water. The retina was loosened from sclera with metal micro-spatula and cut loose at Optic nerve with microscissors in the way that optic nerve head remained attached to the retina. The retina was placed in a perforated sample holder under gently 164 running tap water overnight to remove formalin fixative. The next morning the retina was placed in 25 ml glass beaker with 5 ml of 3% crude trypsin in Tris HCl buffer, pH 7.8 with 0.2M NaF and digested for about 90 min. The inner limiting membrane was removed and the digested retina was placed into a Petri dish containing distilled water using a glass pipette as a transfer tool. The retina was gently brushed with cat whisker hair to dislodge neural elements. The vessel network was cleaned three times in pure distilled water, transferred onto mounting slide, and stained with hematoxylin and periodic acid-Schiff. 3.5. Histological Assessment of Retinal Capillaries The mid-retina areas along each major vessel of the vascular bed were systematically investigated (6-8 fields per retina) to quantify the acellular capillaries at 250x magnification (~0.32 mm2 per field). A capillary was counted as acellular if it had no nuclei from junction to junction and comprises at least 20% of the diameter of surrounding capillaries. 3.6. Simultaneous Extraction of Proteins and RNA The method was performed as previously described (30). Each retina was homogenized in 1 mL of TRIzol reagent. Then 200 pl Chloroform was added, the sample was covered tightly, shacked vigorously for 15 seconds and let it stay at room temperature for 2-3 minutes; then centrifuge at 13,000 g for 15 minutes at 4°C. Phases separation was 165 obtained: the aqueous phase (RNA), the red organic phase (protein), the interphase (DNA). Any residual DNA was precipitated from the organic phase with 100% ethanol (300 pl), followed by a 2000g centrifugation for 5 minutes at 4 °C. The organic phase was resolved in 14 mL of wash buffer (31.25mmol/L Tris-HCI, pH 6.8, 0.1% SDS (sodium dodecyl sulfate)). After centrifugation at 4000g for 5 minutes at 23 °C, the upper phase and wash buffer was transferred to a 5KD MWCO Amicon -15 (Millipore) ultrafiltration tube. The Amicon -15 (Millipore) ultrafiltration tube was centrifuged at 3000g for 20 minutes at 23 °C. The filtrate was discarded and the same wash step was performed 3 times with 14 mL of new wash buffer added each time. The protein solution (approx 250 pl retentate) was transferred to an eppendorf tube; 5 pl of protease inhibitor cocktail was added. The sample was stored in -80 °C until analysis. 3.7. Quantitative Real Time-Polymerase Chain Reaction (qRT-PCR) Total RNA was extracted from rat and mice retinas. Specific primers for each gene are listed in Table 1. Primers were designed using IDT DNA PrimerQuest software (Coralville, IA). First strand cDNA was synthesized using the SuperScript II RNase H- Reverse Transcriptase (Invitrogen Carlsbad, CA). Synthesized cDNA was mixed with 2x SYBR Green PCR Master Mix (Invitrogen Carlsbad, CA).and different sets of gene- specific forward and reverse primers and subjected to real-time PCR quantification using the ABI PRISM 7900 Sequence Detection System (Applied Biosystems). All reactions were performed in triplicate. The relative amounts of mRNAs were calculated by using the comparative CT method (User Bulletin #2, Applied Biosystems). Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRNA. 166 3.8. Western Blot Proteins were extracted from rat retinas using a simultaneous extraction of proteins and RNA method as previously described. Proteins were resolved by NuPAGE Novex 10% Bis-Tris gels, transferred to nitrocellulose membrane and immunobloted using appropriate primary antibodies followed by secondary lRDye infrared secondary antibodies (Invitrogen, Molecular probes, Eugene, OR). Irnmunoreactive bands were visualized and quantified by Odyssey infrared imaging system (Ll-COR Biosciences, Lincoln, NE). 3.9. Immunofluorescence Assay Rat retinal samples were fixed in Zinc Fixative (BD Biosciences, San Jose, CA) for 48 hours, then washed in PBS. Then the samples were blocked 60 minutes with 3% normal serum in 0.3% Triton-x100 in TBS. The primary antibody incubation was overnight at 4°C on rocker. The next day tissue was rinsed in two changes of TBS+Tween20 (TBST), five min each on rocker at room temperature. Secondary fluorescent antibody incubation was three hours at room temperature on rocker. Then tissue was rinsed in three changes of TBST for 10 min each and mounted on slides and covered with a coverslip. 3.10. Rat Model of Retinal Ischemia-Reperfusion Male Sprague-Dawley rats weighing 300 grams were used. All procedures involving the animal models adhered to the ARVO statement for the Use of animals in Ophthalmic and 167 Vision research. Retinal ischemia-reperfusion was created by temporal increase in intraocular pressure to 90 mm Hg as previously described (31). The DHA treated group was on a DHA-enriched diet for 12 days and has also been supplemented via tail vein with 500 nmol/kg DHA one day prior and 250 nmol/kg DHA (32) immediately prior to induction of I/R. The control, no treatment group was on standard rodent diet and subjected to saline solution injection via the tail vein. The retinas were isolated two days after retinal I/R, the time point that corresponds to inflammatory changes similar to the early stage of diabetic retinopathy. 3.11. Slide Preparation and Quantification of the ASMase Staining of the Blood Vessels Rat retinal samples were fixed in Zinc Fixative (BD Biosciences, San Jose, CA) for 48 hours, followed by routine automated vacuum infiltrating tissue processing to paraffin block. Zinc fixed paraffin blocks were sectioned on a rotary microtome at 4 — 5 microns and placed on 3-aminoalkylethoxysilane coated slides, dried overnight at 56°C. Slides following de—paraffinization and hydration to distilled water were placed in TBST (Scytek Labs — Logan, UT) to allow for pH adjustment to 7.4. All slides were stained on a Dako Autostainer (Dako North America — Carpinteria, CA) with TBST rinses between each staining reagent. Non-specific protein block utilized normal serum from the host of the secondary antibody for 30 minutes (Vector Labs — Burlingarne, CA). Endogenous biotin block in Avidin D (Vector) / d-Biotin (Sigma — St. Louis, MO) was used for 15 minutes. Primary antibodies for ASMase and caveolin-1 were applied at optimized 168 dilutions in normal antibody diluent (Scytek) for 1 hour and then biotinylated anti-Host species IgG H+L diluted to l lug/m1 (Vector) in NAD was applied for 30 minutes. R.T.U. Vectastain® Elite ABC Reagent (Vector) was used next for 30 minutes. The reaction was completed with Vector Nova Red peroxidase chromogen (Vector) for— 15 minutes. Slides were then removed from the auto-stainer; counterstained with Gill 2 Hernatoxylin, differentiated, dehydrated, cleared and coverslipped with synthetic mounting medium. The pictures were obtained using a Micropublisher 3.3 Megapixel Color Digital Camera and the amount of staining was quantified using a MetaMorph imaging system (Molecular Devices, Downingtown, PA) 3.12. Statistical Analysis Data are expressed as the mean i SEM for gene expression measurements and mean i SD for western blot quantification and acellular capillary quantification. Factorial ANOVA with post hoc Tukey test (GraphPad PrismS, GraphPad Software, San Diego, CA) was used for comparing the data obtained from multiple independent samples. T-test was used to compare data obtained from two independent measurements. Significance was established at P<0.05. 169 4. Results 4.1. DHA Enriched Diet Prevents the Upregulation of Retinal ASMase in One Month Diabetic Rat Model The natural history of diabetic retinopathy has been divided into a very early pro- inflarnmatory stage and a later, proliferative stage. We assessed the very early stage of diabetic retinopathy in rats affected by type 1 diabetes for a one month period of time (Figure 4.1, 4.2 and 4.3). To investigate whether DHA has a beneficial effect on DR, diabetic rats were subjected to either a soybean oil diet (standard rodent diet) or fish oil (DHA) enriched diet for the duration of the study. The control rats were fed a soybean oil diet. As activation of SMases is an early critical step in inflammatory signaling, we first determined the effect of diabetes on SMases. Interestingly, we found that ASMase gene expression (Figure 4.1) and protein level (Figure 4.3 A,B) was dramatically increased in retinas isolated from diabetic rats on soybean oil diet compared to control, healthy rats on the same diet. Moreover, a DHA (fish oi1)-enriched diet prevented the upregulation of ASMase gene expression (Figure 4.1) and protein levels (Figure 4.3 A,B) in the diabetic rat retina. We found no effect of diabetes or the DHA enriched diet on NSMase gene expression (Figure 4.1), strongly suggesting that ASMase rather than NSMase might play a more important role in mediating vascular and retinal inflammation in the early stage of diabetic retinopathy. 170 4.2. DHA Enriched Diet Prevents the Upregulation of Retinal Proinflammatory/Angiogenic Mediators in One Month Diabetic Rat Model At this initial stage of DR we also found a dramatic upregulation of proinflammatory and pro-angiogenic mediator gene expression, as interleukin 1B (IL-1 [3), IL-6 and vascular endothelial growth factor (VEGF) gene expression was increased in the retinas of diabetic rats fed a soybean oil diet (Figure 4.2). VEGF protein level (Figure 4.3 A,C) was also increased two fold in the diabetic retina from rats fed a soybean oil diet compared to control retina from rats on the same diet. Intercellular adhesion molecule 1 (ICAM-l) plays a critical role in mediating leukocyte adherence to endothelial walls, leukostasis, and endothelial cell injury and death. ICAM-l gene expression (Figure 4.2) was increased three fold in the retinas of diabetic rats fed a soybean oil diet when compared to non—diabetic controls. More importantly, rats fed a DHA (fish oil)- enriched diet did not manifest the diabetes induced upregulation of ICAM-1, VEGF, IL-lB and IL- 6 gene expression (Figure 4.2), as well as VEGF protein level (Figure 4.3 A,C), strongly demonstrating a DHA protective effect against the development of early inflammation in the diabetic retina. 4.3. DHA Enriched Diet Prevents the Upregulation of Retinal ASMase in Nine Months Diabetic Rat Model We next investigated the effect of long-tenn diabetes and a DHA-enriched diet on sphingomyelinase expression in the retina. We found significant upregulation of retinal ASMase gene expression nine months, after induction of diabetes in rats. Interestingly, 171 there was no change in NSMase gene expression between control rats fed a soybean oil diet and diabetic rats fed either a soybean oil or DHA rich diet (Figure 4.4). These results suggest that ASMase rather than NSMase may play the more important role in mediating retinal inflammation and dysfunction in nine-month old diabetic rats. Moreover, a DHA- enriched diet significantly prevented diabetes-induced upregulation of ASMase gene expression (Figure 4.4) and protein levels (Figure 4.5 A,B). 4.4. DHA Enriched Diet Prevents the Upregulation of Retinal Proinflammatory/Angiogenic Mediators in Nine Months Diabetic Rat Model Pro-inflammatory and pro-angiogenic molecules, IL-IB and VEGF, were significantly increased in the nine month-old diabetic rat retinas. Feeding diabetic rats a DHA enriched diet prevented upregulation of IL-lB and VEGF gene expression (Figure 4.6). We found no change in ICAM-l gene expression between the retinas from nine month-old diabetic rats and age matched controls (Figure 4.6). The lack of difference between control and diabetic groups was due to an increase in ICAM-l gene expression with age in control rats, rather than a decrease in ICAM-l level in diabetic animals 4.5. DHA Enriched Diet Prevents Retinal Vessel Loss in Nine Month-Old Diabetic Rat Model To determine whether a DHA enriched diet has a protective effect against the diabetes induced retinal vessel loss, a hallmark for an advanced stage of retinopathy, rat 172 retina vasculature was isolated and the number of degenerated, acellular capillaries was assessed in the retinas of nine month-old diabetic rats subjected to either a DHA-enriched diet or a standard rodent, control diet. Wefound that the number of degenerate, acellular capillaries was dramatically increased in diabetic retinas as compared to control retinas isolated from rats fed soybean oil diet (Figure 4.7 A,B). Moreover, retinas from diabetic rats on a DHA-enriched diet showed significantly fewer acellular capillaries then retinas from diabetic rats fed a soybean oil diet (Figure 4.7 A,B). These data suggest that DHA protects the retinal vasculature against vessel loss, a marker of advanced diabetic retinopathy. 4.6. Upregulation of ASMase and Proinflammatory Cytokines TNFa and lL-lfl in Human Diabetic Retina Next we corroborated our findings in diabetic rats with studies in human diabetic retinas and showed that retinas from diabetic donors exhibited increased ASMase, TNFa and lL-lB mRNA levels when compared to control donors (Figure 4.8). 4.7. ASMase Expression in the Rat Retinal Vasculature Our cell culture data demonstrate that caveolar ASMase expression in endothelial cells rather than other retinal cells plays a key role in cytokine-induced endothelial activation. To determine the distribution pattern of ASMase in vivo, we performed immunohistochemistry of retina flatmounts and cross-sections of the retinal layers in rats. ASMase is an important lysosomal enzyme that is ubiquitously expressed in most tissues. 173 We found basal ASMase expression in all retinal layers (Figure 4.9). Retinal blood vessels, however, exhibited the highest positive staining for ASMase (Figure 4.9) which colocalized with caveolin-1 (Figure 4.9). 4.8. DHA Supplementation Prevents the Upregulation of Vascular ASMase in the Rat Retina Injured by HR Next, we investigated the role of ASMase in the activation and degeneration of retinal vessels using a well established model of accelerated ischemia/reperfusion (I/R) injury that closely resembles diabetic retinopathy (31). ASMase expression was examined in the retinal tissue two days following the I/R injury. Irnmunohistochemistry analysis of retinal cross-sections obtained from uninjured and HR injured rat eyes, showed that I/R injury specifically increased ASMase expression in the retinal vasculature (Figure 4.10) when compared to uninjured eyes, with little or no effect on ASMase expression in other retinal cells (Figure 4.10). As DHA was an effective ASMase inhibitor in our cell culture experiments, we next determined the effect of this w3-PUFA on ASMase expression in the rat retinal tissue isolated from uninjured and I/R injured eyes. DHA supplementation prior to retinal injury specifically prevented ASMase upregulation in the retinal vasculature (Figure 4.10). 174 Table 4.1. Body weight gain and blood glucose of experimental animals with one month of diabetes. Data are mean i SD. Weight gain, g/day Blood Glucose (mM) Control 8.20i1.38 4.8i0.85 Diabetic control diet 4.13:1.6 25:12.5 Diabetic DHA 4.05i1.79 21.7i13.12 supplemented diet Table 4.2. Body weight, blood glucose and HbAlc of experimental animals with nine month of diabetes. Data are mean i SD. Weight at the end of Blood Glucose HbA 1c (%) the study (g) (mM) Control 733.5 i74.24 5.04i0.348 5.3i0.6301 Diabetic control diet 547.8i71.87 23.14i4.21 l6.228i2.649 Diabetic DHA 586.83i88.27 18.15i5.023 13.05i3.117 supplemented diet 175 Table 4.3. Fatty acid composition of the control diet and DHA (fish oil) - enriched diet Fatty acid Control diet (percent of total) DHA supplemented diet mercent of total) 20:5n3 0.00 9.49 18:3n3 7.16 6.46 18:3n6 0.00 0.00 22:6n3 0.00 8.90 16:1n7 0.00 6.73 20:4n6 0.00 0.00 22:5n3 0.00 0.00 18:2n6 45.68 21.88 20:3n6 0.00 0.00 16:0 18.98 26.19 18:1n9 22.88 19.40 18:0 5.31 0.95 n6/n3 ratio Control diet 6.38 DHA supplemented diet 0.88 176 Table 4.4. RT-PCR primers for rat Gene Sense sequence Anti-sense sequence ASMase caactatgggctgaagaagga acagctgactggcacacatt NSMase ttctcgagctttgtccagggttc caggaagtgcttctttggctggt ICAM-l ccaccatcactgtgtattcgtt acggagcagcactactgaga VEGF A gctctcttgggtgcactgg caccacttcatgggctttct IL-l B caaggagagacaagcaacga gtttgggatccacactctcc Cyclophilin cttcttgctggtcttgccattcct tggatggcaagcatgtggtctttg Table 4.5. RT-PCR primers for human Gene Sense sequence Anti-sense sequence ASMase caacctcgggctgaagaa tccaccatgtcatcctcaaaa TNFa tccttcagacacctcaacc aggccccagtttgaattctt IL-1 [3 gggcctcaaggaaaagaatc ttctgcttgagaggtgctga Cyclophilin caagactgagtggttggatgg tggtgatcttcttgctggtct 177 0.04 - * :__c_ 0.035 E 0.03 -- _c_> 3, 0.025 b 0.02 .9 is 0.015 2 g 0.01 l— o _;__.._ ____ ASMase NSMase E1 Control soybean oil I l Diabetes soybean oil Diabetes fish oil (DHA-enriched) Figure 4.1. The effect of a DHA-enriched diet on ASMase retinal gene expression in rats with one month of diabetes. Retinas from rats fed either a soybean oil (standard rodent) diet or DHA (fish oil)-enriched diet, isolated one month afier induction of diabetes were analyzed by RT-PCR for ASMase and NSMase gene expression. Gene expression from retinas isolated from rats subjected to a soybean oil diet (control, white bar; diabetic, black bar) and a DHA-enriched diet (diabetic, gray bar). The relative amounts of mRNAs were calculated by using the comparative CT method. Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRNA. n= 5 to 8 animals; *P<0.05 compared to control soybean oil; #P<0.05 compared to diabetes soybean oil. 178 0.0025 0.12 .5 = * § 0.002 0-1 ' '5 0.08 . 6‘ 0.0015 3 0.06 . '5 0.001 . g 0.04 J m l 1': “-0005 0.02 —3 0 - 0 — .5 0.05 - E 2 0.04 9. o 0.03 b .9 a 0.02 (D g r: 0.01 ,- o IL-1B IL-6 Figure 4.2. The effect of a DHA-enriched diet on retinal inflammatory/angiogenic molecule gene expression in rats with one month of diabetes. Retinas from rats fed either a soybean oil (standard rodent) diet or DHA (fish oil)-enriched diet, isolated one month after induction of diabetes were analyzed by RT-PCR for inflammatory/angiogenic molecule expression. RT-PCR analysis of ICAM-1, VEGF, IL-lB and IL-6 from retinas isolated from rats subjected to a soybean oil diet (control, white bar; diabetic, black bar) and a DHA-rich diet (diabetic, gray bar). The relative amounts of mRNAs were calculated by using the comparative CT method. Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRNA. n= 5 to 8 animals; *P<0.05 compared to control soybean oil; #P<0.05 compared to diabetes soybean oil. 179 Diabetes supplemented Control Diabetes with DHA ' u l , 1 VEGF ----i Tubulin --‘—- B C. ASMase VEGF 1 f- * 1.5 7 .E ‘ :3, i 3 1 _,. l: f E ' 9, T 9 0.5 . °- ; 0 .;__ Figure 4.3. The effect of a DHA-enriched diet on retinal ASMase and VEGF protein expression in rats with one month of diabetes. (A) Immunoblot of ASMase and VEGF protein levels in retinas isolated from control rats fed a soybean oil diet (white bar) and from diabetic rats fed a soybean oil diet (black bar) and DHA-enriched diet (gray bar). Quantitative analysis of the data presented in panel (A) for (B) ASMase and (C) VEGF expression. The results are meaniSD. (n= 4 rats in each group; *P<0.05 compared to control soybean oil; #P<0.05 compared to diabetes soybean oil). 180 0.25 g . * E 0.2 ~ 0. 2 9. o 0.15 b .9 b 0.1 (I) C E? l- 0.05 — 0 .______ ASMase NSMase [J Control soybean oil I Diabetes soybean oil 1 Diabetes fish oil (DHA-enriched) Figure 4.4. The effect of a DHA-enriched diet on retinal ASMase gene expression in rats with nine months of diabetes. Retinas from rats fed either a soybean oil (standard rodent) diet or DHA (fish oil)-enriched diet, isolated nine months after induction of diabetes were analyzed by RT-PCR for ASMase and NSMase gene expression. Gene expression from retinas isolated from rats subjected to a soybean oil diet (control, white bar; diabetic, black bar) and a DHA-rich diet (diabetic, gray bar). The relative amounts of mRNAs were calculated by using the comparative CT method. Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRNA. n= 5 to 8 animals; *P<0.05 compared to control soybean oil; #P<0.05 compared to diabetes soybean oil. 181 A. Diabetes supplemented with Control Diabetes DH A I 11 ll ASMase""" - , “'- Tubulin —— “ B. ASMase Protein/Tubulin ’ El Control soybean oil I I Diabetes soybean oil E E3 Diabetes fish oil (DHA-enriched) L Figure 4.5. The effect of a DHA-enriched diet on retinal ASMase protein level in rats with nine months of diabetes. (A) Immunoblot of ASMase protein levels in retinas isolated from control rats on standard rodent diet (soybean oil diet) (white bar) and diabetic rats on soybean oil diet (black bar) or DHA-enriched diet (gray bar). Equal amounts of protein were added to each lane, as confirmed by tubulin levels. (B) Quantitative analysis of the data presented in panel (A) for ASMase expression. The results are meaniSD. (n=4 rats in each group. *P<0.05 compared to control soybean oil; #P<0.05 compared to diabetes soybean oil). 182 0.0025 012 . 0.0006 c , * % 0.0004 -~; 5 0.0015 - g b 0.0003 7 .3 0.001 I 3 0.0002 s C ‘ I (0 0.0005 - .: 1': 0.0001 & 0 ~ 0 - lL-1B ED Control soybean oil I Diabetes soybean oil 1531 Diabetes fish oil (DHA-enriched) Figure 4.6. The effect of a DHA-enriched diet on retinal inflammatory/angiogenic molecules gene expression in rats with nine months of diabetes. Retinas from rats fed either a soybean oil (standard rodent) diet or DHA (fish oil)-enriched diet, isolated nine months after induction of diabetes were analyzed by RT-PCR for inflammatory/angiogenic molecule expression. RT-PCR analysis of ICAM-1, VEGF and lL-lB from retinas isolated from rats subjected to a soybean oil diet (control, white bar; diabetic, black bar) and a DHA-rich diet (diabetic, gray bar). The relative amounts of mRNAs were calculated by using the comparative CT method. Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRNA. n= 5 to 8 animals; *P<0.05 compared to control; #P<0.05 compared to diabetes soybean oil). 183 Control Diabetes Soybean oil Soybean oil Fish oil 5 ‘ ’ . (control dis!) 0, if: x f *: ‘ ...". 1 I fi ‘15 ‘r-‘\. ‘1 x ‘, .5 \ . 5 fi$ .- ‘ v. \ ., 10,. I 3‘ _‘i 1' \\. - V. ‘ bdhfl' \. R“ t Figure 4.7. The effect of a DHA-enriched diet on retinal vessel loss in rats with nine months of diabetes. Retinas from rats fed either a soybean oil (standard rodent) diet or DHA (fish oil) enriched diet, isolated nine months after induction of diabetes were analyzed by microscopy of mounted retinal vasculature to visualize acellular capillaries. (A) Acellular (degenerated) capillary formation (black arrows) in the retinal vasculature isolated from control and nine months diabetic rats fed a soybean oil diet (n= 8) or a DHA enriched diet (n= 8). 184 16 NE 14 E g 12 - £3 10 —~ % a - 0 LB 6 ~ 2 6 4 O < 2 _ 0 E1 Control soybean oil E . I Diabetes soybean oil E i E [1 Diabetes fish oil (DHA-enriched) ‘ Figure 4.7 (continued). The effect of a DHA-enriched diet on retinal vessel loss in rats with nine months of diabetes. (B) Quantification of the total number of acellular capillaries (average per field, 11: 8 animals per group). At least eight fields of retina were counted in duplicates by two independent investigators, *P<0.05 compared to control; #P<0.05 compared to diabetes soybean oil red bar = 550nm; black bar = 1 10m. 185 ASMase TNFa C 0.03 — . 0.0015 — =72 .. 3 3. 0.02 0.001 b .g . § 0.01 0.0005 g s o n..___ 0 j __ _ C. IL-1B 5 0.004 a .. i 0.003 f 0 ; E1 0.002 ‘j 5 0.001 )— Figure 4.8. Changes in retinal gene expression in control and diabetic human retina. RT-PCR analysis of (A) ASMase, (B) TNFa and (C) IL-lB from retinas isolated from postmortem human control (white bars) and diabetic eyes (black bars). The relative amounts of mRNAs were calculated by using the comparative CT method. Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRNA. *P<0.05 compared to control. 186 ASMase Caveolin-1 Mer ed Figure 4.9. Rat retinal ASMase expression. (A) Colocalization of ASMase and Caveolin-1 in normal rat retina flat mount. (B) ASMase expression in normal rat retina showing ASMase positive staining of retinal vessels (black arrow) and perinuclear lysosomal compartment of retinal ganglion cells (black arrow head). (C) Caveolin-1 expression in normal rat retina showing caveolin-1 positive staining of retinal vessels (black arrow); red bar = 55 um. 187 ASMase staining Uniniured eye I I l/R iniured eye I 11~-.,.-..,.;; II ‘ I $ . ”filyJ'I ‘ ‘3 ’ . . Secondary antibody control , Uninjured eye I I l/R injured eye I ‘ * ~ ~ ' he :1- i . 5‘5 0.. ......f ‘3‘ 0"”. " ' "J., . ‘ v .0 . I 8'. o Figure 4.10. Retinal vascular ASMase expression after retinal I/R injury in rats. (A) Specific increase in ASMase expression in the rat retinal vessels (black arrow) from I/R injured eye (A, right) compared to control (A, left). (B) Secondary antibody control for (A). 188 C: ASMase staining I Uninjured eye I I l/R injured eye I a“ €8.43. - am} ' .5 .1 .xi ”min! f 9 .‘3f2’1'v’ Figure 4.10 (continued). Retinal vascular ASMase expression after retinal I/R injury in rats. DHA supplementation prevents the increase in vascular ASM expression in UK injured eye (C, right). (D) Secondary antibody control for (C). 189 Retinal vessels ., 0.025 -— 11 C . D gag 0.02 § ‘61 , g 2 0.015 ~ on .9 .. U) . gfi 0.01 ‘2 “"273 E 0.005 «3 8 0 Control DHA supplementation F. Neuroretina 1:3 0.025 - 3 1 on 2 .§% 0.02 r E w w '8 0.015 ‘3 a - 9% 0.01 - ‘2 a g 0.005 -« C 0 Control DHA supplementation Figure 4.10 (continued). Retinal vascular ASMase expression after retinal I/R injury in rats. Quantification of ASMase staining in retinal vessels (E) and neuroretina (F). The data is presented from five animals in each group, 11-17 retinal vessels per group; *P<0.05 compared to all other groups. Red bar = 55 um, black bar = 220m. 190 5. Discussion Early stage diabetic retinopathy is regarded as a low-grade chronic inflammatory disease. Inflammation is part of the body's normal response to infection and injury; however, when it happens in an uncontrolled or inappropriate manner it can induce excessive damage to the host tissue and disease may ensue. Such uncontrolled or inappropriate inflammatory responses also occur in diabetic retina and are characterized by increased expression of adhesion molecules on the vascular endothelium and leukocyte surfaces, sequestration of leukocytes to sites where they are not commonly found, production of inflammatory mediators and ultimately damage to the host tissue (2). The molecular mechanisms that lead to a persistent inflammation in the diabetic retina are not well defined, but it seems that hyperglycemia and dyslipidemia, two metabolical characteristics of diabetes mellitus, play an important role in the inflammatory process. Various biochemical pathways have been proposed to represent the link between hyperglycemia and diabetic microangiopathy. Among these pathways, polyol accumulation (33-34), oxidative stress and formation of reactive oxygen species (ROS) (35), increase activity of various PKC isoforms (36-37), and glycation of key proteins that lead to formation of advanced glycation end products (AGES) (38-40) are metabolic pathways that have received much attention. Numerous studies have also demonstrated that AGES production(41), PKCS activation(42), and ROS generation(43), have the ability to activate sphingomyelinases. ASMase and NSMase are considered major candidates for mediating stress-induced cell injury and death; however, these enzymes have individual requirements for their activation and regulation. ASMase functions best 191 at a pH of 4.5-5.5, whereas optimal NSMase activation occurs at pH 7.4, and requires magnesium or manganese (44). Diabetic hyperglycemia and hypoxemia induced local retinal acidosis along with diabetes-related hypomagnesaemia (45) and this may downplay the role of NSMase in diabetic retinopathy. Indeed, our study demonstrates that ASMase rather than NSMase could represent a key player in mediating vascular damage in diabetic retinopathy. Dyslipidemia associated with diabetes has also been implicated in promoting the inflammatory status in the retina. Along with DCCT/EDIC findings for a strong association between dyslipidemia and diabetic retinopathy, our recent study has demonstrated a significant decrease in total retinal (03 PUFAS, and especially DHA, that was tightly coupled with inflammatory status in the diabetic retina (20). At the initial stage of diabetic retinopathy, we also found in this study a dramatic increase in retinal gene expression of the pro-inflammatory and pro-angiogenic mediators, IL-10 and VEGF (Figure 4.2 and Figure 4.3 A,C). The involvement of IL-IB in pathogenesis of diabetic retinopathy has been recently defined in diabetic mice (46). Inhibition of caspase-1, the enzyme involved in production of IL-10 cytokine or inhibition of IL-10 signaling using IL-1|3 receptor deficient mice protected the mice from development of retinal pathology over seven months duration of diabetes (46). VEGF is a key molecule that plays a well- recognized role in promoting vascular permeability (47) and pathological ocular neovascularization (48). VEGF expression is mainly regulated by hypoxic stimulus; yet there is evidence that it also accumulates in the retina early in diabetes, before any retinal hypoxia is present (49-51) and our results are in accordance with these studies. 192 Inflammatory mediators can promote the activation and degeneration of capillary endothelial cells with subsequent profound impact on vascular permeability and viability. ICAM-l plays a critical role in mediating leukocytes adherence to endothelial walls, leukostasis, increased movement of leukocytes from the bloodstream into the surrounding tissue and endothelial cell injury and death. Pro-inflammatory cytokines, including IL-10 and VEGF have a pronounced effect on upregulation of ICAM-1 expression in the retina. We found in our study that ICAM-l gene expression was increased three fold in the retina from diabetic rats on a standard diet when compared to control (Figure 4.2). ASMase is regarded as an initial pivotal responder in diverse cellular processes of physiologic and pathologic importance. ASMase induction by various cytokine and stress stimuli has been reported to play an essential role in cytokine-mediated inflammation, apoptosis, cellular differentiation and cellular senescence (52-53). At the initial stage of DR, we show that ASMase gene expression and protein levels were significantly increased in the diabetic retina. The increased expression of many inflammatory proteins is regulated at the gene transcription level through the activation of inflammatory transcription factors, including nuclear factor kappa B (N F-KB), activator protein 1 (AP-1), specificity protein 1 (SP1) and other members of the nuclear receptor family (54-55). Analysis of the ASMase promoter region identified several promoter elements essential for basal and inducible ASMase gene expression; among these, SP-l, AP-l and AP-2 are mostly characterized and a concerted action of these transcription factors appears to be essential in increased ASMase expression (56-57). Interestingly, the upregulation of ASMase expression in diabetic retina could potentiate the cytokine pleiotropic signaling properties in retinal inflammation. Moreover, I/R injury-mediated upregulation of ASMase expression specifically in the retinal 193 vasculature may have a profound effect on cytokine induced inflammatory process in the vascular endothelium, which ultimately results in endothelial cells activation, dysfunction and death. Omega-3 PUFAS, and particularly DHA are regarded as potent anti-inflammatory agents and they may be of therapeutic benefit in a diversity of acute and chronic inflammatory conditions (58). Our findings support the anti-inflammatory effect of DHA and show that DHA supplementation decreases the expression of inflammatory mediators and ICAM-1 in diabetic retina. More importantly, DHA decreases ASMase expression in the diabetic retina and I/R injured vasculature, and this in vivo study recapitulates our observations in in vitro settings where ASMase downregulation by DHA or ASMase gene silencing had significant inhibitory effects on cytokine-induced upregulation of adhesion molecules expression in retinal endothelium. Furthermore, DHA supplementation conferred significant protection against the development of acellular capillaries, the hallmark of late stage diabetic retinopathy. In conclusion, we established the protective effect of DHA in in vivo models of diabetic retinopathy. DHA downregulates ASMase expression, prevents retinal inflammation and retinal capillary loss in rats with DR. Moreover, DHA modulates vascular ASMase expression in the l/R injured rat retina. These results demonstrate that ASMase may play a pivotal role in retinal inflammation and vascular degeneration in type 1 diabetic rat animal model. Consequently, in the following chapter, we directly address the role of ASMase in retinal vascular microangiopathy using in vivo accelerated models of retinopathy, including retinal ischemia-reperfusion and oxygen-induced retinopathy that were induced in an ASMase deficient (ASMase 4') mouse. 194 6. References l. 10. ll. Sinclair, A.J., Bayer, A.J., Girling, A.J., and Woodhouse, K.W. 2000. Older adults, diabetes mellitus and visual acuity: a community-based case-control study. Age Ageing 29:335-339. Kern, TS. 2007. Contributions of inflammatory processes to the development of the early stages of diabetic retinopathy. Exp Diabetes Res 2007:95103. Lee, P., Wang, CC, and Adamis, AP 1998. Ocular neovascularization: an epidemiologic review. Surv Ophthalmol 43:245-269. Meleth, A.D., Agron, E., Chan, C.C., Reed, G.F., Arora, K., Bymes, G., Csaky, K.G., Ferris, F.L., 3rd, and Chew, E.Y. 2005. Serum inflammatory markers in diabetic retinopathy. Invest Ophthalmol Vis Sci 46:4295-4301. Joussen, A.M., Murata, T., Tsujikawa, A., Kirchhof, B., Bursell, S.E., and Adamis, AP. 2001. Leukocyte-mediated endothelial cell injury and death in the diabetic retina. Am J Pathol 158:147-152. McLeod, D.S., Lefer, D.J., Merges, C., and Lutty, GA. 1995. Enhanced expression of intracellular adhesion molecule-1 and P-selectin in the diabetic human retina and choroid. Am J Pathol 147:642-653. Limb, G.A., Chignell, A.H., Green, W., LeRoy, F., and Dumonde, DC. 1996. Distribution of TNF alpha and its reactive vascular adhesion molecules in fibrovascular membranes of proliferative diabetic retinopathy. Br J Ophthalmol 80:168-173. Joussen, A.M., Poulaki, V., Le, M.L., Koizumi, K., Esser, C., Janicki, H., Schraermeyer, U., Kociok, N., Fauser, S., Kirchhof, B., et a1. 2004. A central role for inflammation in the pathogenesis of diabetic retinopathy. Faseb J 18:1450- 1452. Murata, T., Nakagawa, K., Khalil, A., Ishibashi, T., lnomata, H., and Sueishi, K. 1996. The relation between expression of vascular endothelial growth factor and breakdown of the blood-retinal barrier in diabetic rat retinas. Lab Invest 74:819- 825. Adamis, A.P., Miller, J .W., Bemal, M.T., D'Amico, D.J., Folkman, J ., Yeo, T.K., and Yeo, K.T. 1994. Increased vascular endothelial growth factor levels in the vitreous of eyes with proliferative diabetic retinopathy. Am J Ophthalmol 118:445-450. Gustavsson, C., Agardh, C.D., Hagert, P., and Agardh, E. 2008. Inflammatory markers in nondiabetic and diabetic rat retinas exposed to ischemia followed by reperfusion. Retina 28:645-652. 195 12. l3. 14. 15. l6. 17. 18. 19. 20. 21. Liu, P., and Anderson, R.G. 1995. Compartmentalized production of ceramide at the cell surface. J Biol Chem 270:27179-27185. Mathias, S., Younes, A., Kan, C.C., Orlow, 1., Joseph, C., and Kolesnick, RN. 1993. Activation of the sphingomyelin signaling pathway in intact EL4 cells and in a cell-free system by IL-1 beta. Science 259:519-522. Yang, Z., Costanzo, M., Golde, D.W., and Kolesnick, RN. 1993. Tumor necrosis factor activation of the sphingomyelin pathway signals nuclear factor kappa B translocation in intact HL-60 cells. J Biol Chem 268:20520-20523. Marathe, S., Schissel, S.L., Yellin, M.J., Beatini, N., Mintzer, R., Williams, K.J., and Tabas, I. 1998. Human vascular endothelial cells are a rich and regulatable source of secretory sphingomyelinase. Implications for early atherogenesis and ceramide-mediated cell signaling. J Biol Chem 273 :4081-4088. K0, Y.G., Lee, J.S., Kang, Y.S., Ahn, J.H., and Seo, J.S. 1999. TNF-alpha- mediated apoptosis is initiated in caveolae-like domains. J Immunol 162:7217- 7223. Martin, M.U., and Wesche, H. 2002. Summary and comparison of the signaling mechanisms of the Toll/interleukin-l receptor family. Biochim Biophys Acta 1592:265-280. Wong, M.L., Xie, B., Beatini, N., Phu, P., Marathe, S., Johns, A., Gold, P.W., Hirsch, E., Williams, K.J., Licinio, J ., et al. 2000. Acute systemic inflammation up-regulates secretory sphingomyelinase in vivo: 3 possible link between inflammatory cytokines and atherogenesis. Proc Natl Acad Sci U S A 97:8681- 8686. . Busik, J .V., Mohr, S., and Grant, MB. 2008. Hyperglycemia-induced reactive oxygen species toxicity to endothelial cells is dependent on paracrine mediators. Diabetes 57:1952-1965. Tikhonenko, M., Lydic, T.A., Wang, Y., Chen, W., Opreanu, M., Sochacki, A., McSorley, K.M., Renis, R.L., Kern, T., Jump, D.B., et al. 2009. Remodeling of Retinal Fatty Acids in an Animal Model of Diabetes: a Decrease in Long Chain Polyunsaturated Fatty Acids is Associated with a Decrease in Fatty Acid Elongases Elov12 and Elovl4. Diabetes. Connor, K.M., SanGiovanni, J.P., Lofqvist, C., Aderman, C.M., Chen, J., Higuchi, A., Hong, S., Pravda, E.A., Majchrzak, S., Carper, D., et al. 2007. Increased dietary intake of omega-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nat Med 13:868-873. 196 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. Chen, W., Esselman, W.J., Jump, DB, and Busik, J .V. 2005. Anti-inflammatory effect of docosahexaenoic acid on cytokine-induced adhesion molecule expression in human retina] vascular endothelial cells. Invest Ophthalmol Vis Sci 46:4342-4347. Opreanu, M., Lydic, T.A., Reid, G.E., McSorley, K.M., Esselman, W.J., and Busik, J. 2010. Docosahexaenoic Acid Inhibits Cytokine Signaling in Human Retinal Endothelial Cells by Downregulating Sphingomyelinases. Invest Ophthalmol Vis Sci. Futterman, S., and Kupfer, C. 1968. The fatty acid composition of the retinal vasculature of normal and diabetic human eyes. Invest Ophthalmol 7: 1 05-108. Simopoulos, AP. 2003. Importance of the ratio of omega-6/omega-3 essential fatty acids: evolutionary aspects. World Rev Nutr Diet 92:1-22. de Lorgeril, M., and Salen, P. 2003. Dietary prevention of coronary heart disease: focus on omega-6/omega-3 essential fatty acid balance. World Rev Nutr Diet 92:57-73. Cleland, L.G., James, M.J., and Proudrnan, SM. 2003. Omega-6/omega-3 fatty acids and arthritis. World Rev Nutr Diet 92:152-168. Zheng, L., Szabo, C., and Kern, T .S. 2004. Poly(ADP-ribose) polymerase is involved in the development of diabetic retinopathy via regulation of nuclear factor-kappaB. Diabetes 53:2960—2967. Kern, T.S., Tang, J., Mizutani, M., Kowluru, R.A., Nagaraj, R.H., Romeo, G., Podesta, F., and Lorenzi, M. 2000. Response of capillary cell death to aminoguanidine predicts the development of retinopathy: comparison of diabetes and galactosemia. Invest Ophthalmol Vis Sci 41 :3972-3978. Agardh, E., Gustavsson, C., Hagert, P., Nilsson, M., and Agardh, CD. 2006. Modifying a standard method allows simultaneous extraction of RNA and protein, enabling detection of enzymes in the rat retina with low expressions and protein levels. Metabolism 55:168-174. Zheng, L., Gong, B., Hatala, D.A., and Kern, TS. 2007. Retinal ischemia and reperfusion causes capillary degeneration: similarities to diabetes. Invest Ophthalmol Vis Sci 48:361-367. Huang, W.L., King, V.R., Curran, O.E., Dyall, S.C., Ward, R.B., Lal, N., Priestley, J .V., and Michael-Titus, AT. 2007. A combination of intravenous and dietary docosahexaenoic acid significantly improves outcome after spinal cord injury. Brain 13023004-3019. 197 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. Engerman, R.L., and Kern, TS. 1984. Experimental galactosemia produces diabetic-like retinopathy. Diabetes 33 :97-1 00. Frank, R.N., Keim, R.J., Kennedy, A., and Frank, K.W. 1983. Galactose-induced retinal capillary basement membrane thickening: prevention by Sorbinil. Invest Ophthalmol Vis Sci 24:1519-1524. Giugliano, D., Ceriello, A., and Paolisso, G. 1996. Oxidative stress and diabetic vascular complications. Diabetes Care 19:257-267. Shiba, T., Inoguchi, T., Sportsman, J .R., Heath, W.F., Bursell, S., and King, G.L. 1993. Correlation of diacyl glycerol level and protein kinase C activity in rat retina to retinal circulation. Am J Physiol 265:E783-793. Ways, D.K., and Sheetz, M.J. 2000. The role of protein kinase C in the development of the complications of diabetes. Vitam Horm 60: 149-193. Brownlee, M. 1992. Glycation products and the pathogenesis of diabetic complications. Diabetes Care 15: 1 835-1 843. Brownlee, M., Vlassara, H., and Cerarni, A. 1984. Nonenzymatic glycosylation and the pathogenesis of diabetic complications. Ann Intern Med 101 2527-53 7. Wautier, J .L., and Guillausseau, P.J. 2001. Advanced glycation end products, their receptors and diabetic angiopathy. Diabetes Metab 27:535-542. Patschan, S., Chen, J ., Polotskaia, A., Mendelev, N., Cheng, J ., Patschan, D., and Goligorsky, MS. 2008. Lipid mediators of autophagy in stress-induced premature senescence of endothelial cells. Am J Physiol Heart Circ Physiol 294:H1119- 1129. Zeidan, Y.H., Wu, B.X., Jenkins, R.W., Obeid, L.M., and Hannun, Y.A. 2008. A novel role for protein kinase Cdelta-mediated phosphorylation of acid sphingomyelinase in UV light-induced mitochondrial injury. F aseb J 22: 1 83-193. Dumitru, C.A., Zhang, Y., Li, X., and Gulbins, E. 2007. Ceramide: a novel player in reactive oxygen species-induced signaling? Antioxid Redox Signal 921535- 1540. Marchesini, N., and Hannun, Y.A. 2004. Acid and neutral sphingomyelinases: roles and mechanisms of regulation. Biochem Cell Biol 82:27-44. Pham, P.C., Pham, P.M., Pham, S.V., Miller, J.M., and Pham, RT. 2007. Hypomagnesemia in patients with type 2 diabetes. Clin J Am Soc Nephrol 2:366- 373. 198 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. Vincent, J .A., and Mohr, S. 2007. Inhibition of caspase-l/interleukin-lbeta signaling prevents degeneration of retinal capillaries in diabetes and galactosemia. Diabetes 56:224-230. Senger, D.R., Connolly, D.T., Van de Water, L., Feder, J., and Dvorak, HF. 1990. Purification and NH2-terminal amino acid sequence of guinea pig tumor- secreted vascular permeability factor. Cancer Res 50:1774-1778. Ng, E.W., and Adamis, AP. 2005. Targeting angiogenesis, the underlying disorder in neovascular age-related macular degeneration. Can J Ophthalmol 40:352-368. Gerhardinger, C., Brown, L.F., Roy, S., Mizutani, M., Zucker, C.L., and Lorenzi, M. 1998. Expression of vascular endothelial growth factor in the human retina and in nonproliferative diabetic retinopathy. Am J Pathol 152:1453-1462. Segawa, Y., Shirao, Y., Yamagishi, S., Higashide, T., Kobayashi, M., Katsuno, K., lyobe, A., Harada, H., Sato, F., Miyata, H., et al. 1998. Upregulation of retinal vascular endothelial growth factor mRNAs in spontaneously diabetic rats without ophthalmoscopic retinopathy. A possible participation of advanced glycation end products in the development of the early phase of diabetic retinopathy. Ophthalmic Res 30:333-339. Sone, H., Kawakami, Y., Okuda, Y., Sekine, Y., Honmura, S., Matsuo, K., Segawa, T., Suzuki, H., and Yamashita, K. 1997. Ocular vascular endothelial growth factor levels in diabetic rats are elevated before observable retinal proliferative changes. Diabetologia 40:726-730. Hannun, Y.A. 1997. Sphingolipid second messengers: tumor suppressor lipids. Adv Exp Med Biol 400A:305-312. Kolesnick, RN, and Kronke, M. 1998. Regulation of ceramide production and apoptosis. Annu Rev Physiol 60:643-665. Rahman, I. 2002. Oxidative stress, transcription factors and chromatin remodelling in lung inflammation. Biochem Pharmacol 64:93 5-942. Winyard, P.G., and Blake, DR. 1997. Antioxidants, redox-regulated transcription factors, and inflammation. Adv Pharmacol 38:403—421. Langrnann, T., Buechler, C., Ries, S., Schaeffler, A., Aslanidis, C., Schuierer, M., Weller, M., Sandhoff, K., de Jong, P.J., and Schmitz, G. 1999. Transcription factors Spl and AP-2 mediate induction of acid sphingomyelinase during monocytic differentiation. J Lipid Res 40:870-880. 199 57. 58. Schuchman, E.H., Levran, O., Pereira, L.V., and Desnick, R.J. 1992. Structural organization and complete nucleotide sequence of the gene encoding human acid sphingomyelinase (SMPDI). Genomics 12:197-205. Calder, P.C. 2006. n-3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr 83:15058-15198. 200 Chapter V Genetic Deficiency of Acid Sphingomyelinase Prevents Upregulation of Inflammatory/Angiogenic Mediators, Vessel Loss and Neovascularization in the Retina of Mice with Microangiopathy Lesions I . Abstract Acid sphingomyelinase (ASMase) is a key candidate for stress-mediated cellular activation and death. We have previously shown that ASMase was upregulated in diabetic rat retina and closely correlated with increased inflammatory/pro-angiogenic molecule levels in the same tissue. More importantly, ASMase expression specifically increased in the rat retinal vasculature injured by ischemia/reperfusion when compared to the non-injured eye. To directly investigate the role of ASMase in endothelial cell activation and death and development of retinal acellular capillaries, we used accelerated models of retinopathy by inducing ischemia! reperfusion (I/R) injury and oxygen-induced retinopathy (OIR) in wild type (ASMaseV) or ASMaseT mouse retina. We found that genetic deficiency of ASMase significantly prevented the increase in inflammatory/pro- angiogenic molecules expression in I/R injured retina compared to wild type. Moreover, I/R injured ASMaseT mouse retinas were protected from both vascular degeneration and neovascularization. These results demonstrate that ASMase deficiency has a protective effect on ischemia-driven endothelial cell dysfunction and death, preventing consequent development of degenerated capillaries, as well as neovessel formation in the injured retinas. 201 2. Introduction Sphingomyelin is an important sphingolipid component of all mammalian membranes and consists of a ceramide backbone with a phosphorylcholine moiety. ASMase represents a key regulatory enzyme in sphingolipid metabolism, catalyzing the hydrolysis of sphingomyelin to the bioactive lipid ceramide. ASMase major role in cell signaling is closely linked to its ability to reorganize the plasma membrane where sphingomyelin hydrolysis into ceramide facilitates the formation and coalescence of plasma membrane lipid microdomains. These ceramide enriched microdomains are believed to increase the local protein density, including receptors that often require oligomerization for their activation or facilitate protein-protein interaction (1). Receptor oligomerization and adaptor proteins recruitment appear to be a requirement for TNFa and IL-1 Signalosome organization and intracellular inflammatory signaling (2-3). Until the middle of the last decade, interest in ASMase biological role was restricted primarily to researchers studying Niernann-Pick disease, a recessively inherited lysosomal storage disease due to ASMase deficiency. At the same time, ASMase knockout mice were generated and become available for research use (4). The use of these ASMase knockout mice opened new avenues of research since these animals were initially found to be resistant to radiation (5). Subsequently, there has been an advancing crescendo of studies that have begun to shed significant light on our understanding on ASMase involvement in cell survival and pathophysiology of cancer, diabetes, sepsis, cardiovascular, pulmonary and neurological diseases (6). However, the role of ASMase in 202 the pathology of diabetic retinal microangiopathy has not been address so far and represents the purpose of this study. Vascular endothelium represents a very rich source of ASMase (5, 7) and, as presented in the previous chapter, retinal vasculature exhibits significant positive staining for ASMase when compared to other retinal tissues. Elevated ASMase concentration in the vasculature makes it an excellent candidate for locally mediation of inflammatory and cell death, signaling pathways that would ultimately result in vascular degeneration. In this study, we directly tested the hypothesis that ASMase genetic deficiency may be protective against the endothelial activation, injury and death, preventing the retinal vessels loss and subsequent pathological angiogenesis. We have modeled retinopathy in an ASMase knockout (ASMase '/') mouse model of Niemann-Pick disease by using retinal ischemia/reperfusion (I/R) and oxygen-induced retinopathy (OIR), both representing accelerated models of retinopathy; while the first model permits us to investigate the formation of acelullar capillaries, the last one allows us the assessment of neovessels formation. The ischemia-reperfusion model induces retinal capillary degeneration, is well established and has close similarities to diabetes induced retinal vascular microangiopathy (8). Ischemia and reperfusion lead to individual effects on tissues related to metabolically disregulation induced by both hypoxia and reoxygenation. The arrest of blood supply in the retina results in cell death via both necrosis and apoptosis. Hypoxia leads to disruption of cellular energy metabolism and initiates a harmful cascade of events (9); induction of cytokine production, generation of reactive oxygen species (ROS), degradation of the anti-oxidant system and extracellular buildup of glutamate are 203 the main changes secondary to ischemia (9-14). The reoxygenation process, although effective in decreasing the ischemic damage, promotes additional cell injury and apoptosis (15) and the reoxygenation-induced cell damage is largely due to generation of reactive oxygen species (ROS) (16), increased levels of extracellular neurotransmitters and waste products that damage previously unharmed cells when being reoxidized (9, 17- 18). Ocular angiogenesis in response to tissue ischemia is commonly seen in various clinical conditions, including diabetic retinopathy, retinopathy of prematurity and retinal vein occlusion (19-20). The oxygen-induced retinopathy model uses a basic premise by which exposure to hyperoxia during early development of retinal vasculature results in the reduction of normal retinal vascular development and the degeneration of newly developed vessels. Removal of the hyperoxic stimulus renders a relative ischemic state in the retina, since the retina is no longer immersed in the high oxygen setting and lacks its normal vasculature. This ischemic situation sets off a rapid pathologic angiogenic process with formation of neovascular tufts which protrude out of the normal retinal vascular plexuses into the vitreal cavity (21 ). In the previous chapter, we demonstrated that ASMase gene expression and protein level are significantly upregulated in diabetic rat retinas that exhibited an inflammatory status and microangiopathy lesions; importantly, ASMase was significantly increased in the vascular rat retina with I/R injury. ASMase is considered to be a first responder in mediating signaling processes via a specific receptor and genetic ASMase knockout mice exhibit protection from stress-induced cell death in various organs and systems (6, 22-23). Therefore, in this study we hypothesized that the ASMase genetic 204 deficiency will render protection against upregulation of proinflammatory/angiogenic mediators, vessel loss and pathological angiogenesis in the retina injured by ischemia- reperfusion and hyperoxic stimulus, respectively. 205 3. Materials and Methods 3.1. Reagents NuPAGE Novex 10% Bis- Tris gels, Platinum SYBR Green qPCR SuperMix-UDG w/ROX were purchased from Invitrogen (Carlsbad, CA). Lyophilized pancreatic elastase was purchased from Calbiochem (San Diego, CA). RT-PCR primers from IDT DNA Technologies (Coralville, IA) (Table 1) were used. ASM antibody was a generous gift from Dr. Richard Kolesnick. High Pure PCR Template Preparation Kit (Roche Applied Science, Mannheim, Germany) was used for DNA isolation. DNase, RNase free water, dNTPs, MgCl, Taq DNA Polymerase, Trackit Cyan/Yellow Buffer, Tracklt lkb Plus DNA ladder were purchase from Invitrogen, (Carlsbad, CA). 3.2 Genotyping Transgenic Mice by PCR and Southemblot 3.2.1. DNA isolation from mouse tail. 0.2 to 0.5 cm of the mouse tail was used for DNA isolation using a High Pure PCR Template Preparation Kit (Roche Applied Science, Mannheim, Germany). The mouse tail was added to a1.5 mL nuclease-free microcentrifuge tube, along with 200 uL Tissue Lysis Buffer and 20 uL Proteinase K (reconstituted). The mixture was incubated overnight at 55 °C until the tissue was completely digested. Using a 1 mL disposable syringe without needle, the tail sample was lysed by shearing process. Then 200 uL of Binding Buffer and 100 uL of isopropanol were added to the tube, mixed well and subjected to centrifugation for 5 minutes at 13,000 x g. Then the liquid sample was pipetted into the upper buffer reservoir of the High Filter Tube that was previously inserted into one Collection tube. The entire High Filter Tube assembly was inserted into a standard table-top centrifuge and centrifuged 1 206 minute at 8,000 x g. After centrifugation, the Filter Tube was removed from the Collection Tube; the flowthrough liquid and the Collection Tube were discarded. The Filter Tube was combined with a new Collection Tube; 500 uL Inhibition Removal Buffer was added to the upper reservoir of the Filter Tube and centrifuged 1 min at 8,000 x g. After centrifugation, the Filter Tube was again removed from the Collection Tube; the flowthrough liquid and the Collection Tube were discarded. The Filter Tube was combined with a new Collection Tube; 500 uL Wash Buffer was added to the upper reservoir of the Filter Tube and centrifuged 1 min at 8,000 x g. This final step was repeated twice. To elute the DNA, the Filter Tube was inserted into a clean, sterile 1.5 mL microcentrifuge tube; 200 uL prewarmed Elution Buffer was added to the upper reservoir of the filter tube and centrifuged 1 min at 8,000 x g. The eluted DNA was stored at -20 0C for further analysis. 3.2.2. PCR amplification of the DNA sample. The following reagents were added to an RNase, DNase fi'ee microcentrifuge tube (a total volume of 25 uL): DNase, RNase fi‘ee water 16 uL, 10x buffer 2.5 uL, dNTPs (10mM) 1 11L, MgCl (150mM) 1.5 11L, Primer Mix for ASMase (0.1 mM) 1.5 uL, Taq DNA Polymerase (Invitrogen, (Carlsbad, CA)) 0.5 uL, DNA 2 uL. The following primers for ASMase have been used: GGCTACCCGT GATATTGC, AGCCGTGTCCTCTTCCTTAC, CGAGACTGTI‘GCCAGACATC. The PCR mix underwent the following cycling conditions: 94 °C for 3 minutes, followed by a sequence (94 0C for 15 seconds, 64 °C for 30 seconds and 68 0C for 90 seconds) that was repeated 35 times and then 72 °C for 7 minutes. 3.2.3. Southemblot identification of ASMase mutant allele. The PCR product (25 pL) was mixed to 2.5 11L of loading buffer (Trackit Cyan/Yellow Buffer, Invitrogen, 207 Carlsbad, CA). 1.5% Agarose in 50 mL TBE buffer, mixed with 5 uL of 10,000x SYBR safe stain was used to cast the gel. The Tracklt lkb Plus DNA ladder was added to the first well. 10 uL of samples to analyze was added to the remaining wells. The ASMase mutant allele was identified at 523 bp, whereas the wild-type allele was localized at 269 bp. 3.3. Mice. All procedures involving the animal models adhered to the ARVO statement for the Use of animals in Ophthalmic and Vision research. Mouse model of retinal ischemia-reperfusion. Two months old male C57BL/6J wild type (ASM+/+) and C57BL/6J ASMase knock-out (ASM'/') mice were anesthetized. The anterior chamber of one eye was cannulated with a 30 gauge needle attached to a line infusing sterile normal saline solution. The intraocular pressure (IOP) was maintained at 80-90 mmHg; this pressure was determined by the height and the volume of the perfusion bag. This resulted in retinal ischemia, as confirmed by whitening of the iris and loss of the red reflex. The other eye of the same animal was used as a control. The duration of ischemia was 90 minutes. After ischemia, the needle was withdrawn, followed by IOP normalization and reperfusion of the retinal vasculature was documented visually. The mice were sacrificed at different times after reperfusion (two days for assessment of inflammatory mediators and seven days for assessment of acellular, degenerated capillaries). Mouse model of oxygen-induced retinopathy (OIR). In the neonatal mouse model of oxygen-induced retinopathy, 7-day-old (P7) C57BL/6J wild type and C57BL/6J acid sphingomyelinase deficient (ASMase'/') mice were placed with their nursing dams in a 75% oxygen atmosphere for 5 days (P7 to P12). Upon return to normal air, these mice develop retinal 208 neovascularization, with peak development occurring 5 days after their return to normoxia. Retinal neovascularization was assessed at P17 as previously described (21, 24). 3.4. Isolation of Retinal Vasculature Mouse retinal vascular beds were isolated by elastase digestion method as previously described (25). The fixed mouse retinas were isolated and rinsed in deionized water as described above. The next day retina was incubated for 15 minutes in an agitating water bath preheated to 37°, in 40 units/ml elastase in 100 mmol/l sodium phosphate buffer with 150 mmol/l sodium chloride and 5.0 mmol/l ethylenediamenetetraacetic acid (EDTA) at pH 6.5. The tissue was washed overnight in 100 mmol/l Tris-HCl (pH 8.5) at room temperature and then transferred to deionized water for removal of the now- loosened vitreous and digested neural elements by gentle agitation using the sides of forceps and the sides and ends of very fine sable brushes as needed. Then the retinas were mounted by flotation in HPLC-grade water over slides. Once air-dried in a dust-free environment, the mounts of the retinal vasculature were stained using the periodic-acid- Schiff reaction and hematoxylin counterstaining. 3.5. Histological Assessment of Retinal Capillaries The mid-retina areas along each major vessel of the vascular bed were systematically investigated (6-8 fields per retina) to quantify the acellular capillaries at 250x magnification (~0.32 mm2 per field). A capillary was counted as acellular if it had no 209 nuclei from junction to junction and comprises at least 20% of the diameter of surrounding capillaries. 3.6. Simultaneous Extraction of Proteins and RNA The method was performed as described in reference (26). The method has been described in detail in chapter IV. 3.7. Quantitative Real Time-Polymerase Chain Reaction (qRT-PCR) Total RNA was extracted from rat and mice retinas. Specific primers for each gene are listed in Table l. The method has been previously described in chaper II. 3.8. Statistical analysis Data are expressed as the mean i SEM for gene expression measurements and mean i SD for western blot quantification and acellular capillary quantification. Factorial ANOVA with post hoc Tukey test (GraphPad PrismS, GraphPad Software, San Diego, CA) was used for comparing the data obtained from multiple independent samples. T-test was used to compare data obtained from two independent measurements. Significance was established at P<0.05. 210 4. Results 4.1. Retinal I/R Induces Upregulation of ASMase Gene Expression in the ASMase”+ Retina Retinas from control and ischemia/reperfusion (l/R) injured eye were isolated from wild type (ASMase+/+) and ASMase genetic deficient (ASMase'f) mice 2 days after induction of the retinal damage. Real time qPCR analysis of ASMase gene expression showed a significant upregulation of ASMase mRNA in wild type retinas injured by I/R (Figure 5.1). Conversely, NSMase gene expression was not significantly increased in the wild type retinas with I/R lesions, but was dramatically elevated in the I/R injured retinas isolated fi'om the ASMaseT mice (Figure 5.2). These results confirmed the role of ASMase as a major candidate in mediating stress-induced cell death. 4.2. ASMase Genetic Deficiency Prevents Upregulation of Cellular Adhesion Molecules in the HR Injured Retina Two days after ischemia-reperfusion, a time point that corresponds to an early stage of diabetic retinopathy, the gene expression of intercellular adhesion molecule 1 (ICAM-l) (Figure 5.3 A) and vascular cell adhesion molecule 1 (VCAM-l) (Figure 5.3 B) was significantly increased in the wild type retinas injured by UK. Conversely, ICAM- 1 and VCAM-l gene expression was significantly decreased in injured retinas isolated form ASMase'f mice when compared to retinas isolated from ASMaseJrl+ I/R injured eye. 211 4.3. ASMase Genetic Deficiency Prevents Upregulation of Inflammatory/ Angiogenic Mediators in the I/R Injured Retina Similarly to adhesion molecules expression, the gene expression of IL-1 [3 (Figure 5.4 A), TNFa (Figure 5.4 B) and VEGF (Figure 5.4 C) was significantly increased in the wild type retinas injured by I/R two days after ischemia-reperfusion. More importantly, ASMaseT mice exhibit IL-lB, TNFa and VEGF gene expression (Figure 5.4 A,B,C) at a normal level in injured retinas compared to the wild type. Therefore, ASMase genetic deficiency emerges as greatly protective against the development of an inflammatory state in the I/R injured retina. 4.4. ASMase Genetic Deficiency Prevents the Development of Acellular Capillaries in the [IR Injured Retina Examination of mouse retinal vasculature in wild-type mice seven days after I/R injury, when capillary degeneration response is greatest (8), showed a dramatic increase in the number of acellular capillaries as compared to control retinas (Figure 5.5 A, C). Furthermore, I/R injured ASMase'/' mouse retinas exhibited significantly fewer acellular capillaries compared to the wild type (Figure 5.5 B, C). These results clearly demonstrate that ASMase deficiency has a protective effect on ischemia-driven endothelial cell dysfunction and death, preventing consequent formation of acellular capillaries. 212 4.5. ASMase Genetic Deficiency Prevents Ocular Neovascularization in OIR Mouse Model The experiments conducted so far investigated the protective role of ASMase deficiency on the development of vascular retinal nonproliferative changes similar to those that develop in diabetic retinopathy. Owing to the lack of an animal model, that properly reproduces the development of proliferative diabetic retinopathy; we next focused our study on an experimental model of ischemic neovascularization involving oxygen-induced retinopathy (OIR). The OIR model allows the assessment of abnormal retinal angiogenesis, which is similar to aberrant new vessel formation in late proliferative stages of diabetic retinopathy. The degree of neovascularization was quantified by counting the number of endothelial cell nuclei present on the vitreal side of the inner limiting membrane (ILM). We found the average number of nuclei extending past the ILM per 6 um cross-section to be 12.1i3.4 SD in hyperoxia-treated ASMaseT mouse eyes, compared to 49.5i12.5 SD in hyperoxia-treated wild type mouse eyes (Figure 5.6). These findings demonstrate that ASMase deficiency prevents the development of pathological angiogenesis in the OIR retina. 213 Table 5.1. RT-PCR mouse primers Gene Sense sequence Anti-sense sequence ASMase NSMase ICAM-l VEGF A 11.40 VCAM-l TNFa Cyclophilin aaccctggctaccgagtttaccaa atgcacactacttcagaagcggga acactatgtggactggcagtggtt tcaccaaagccagcacataggag aagggctgcttccaaacctttgac cccaggtggaggtctactca tctcatgcaccaccatcaagg attcatgtgccagggtggtga tggcctgggtcagattcaagatgt aggcattgagcaccagtccactta tgaggctcgattgttcagctgcta ttacacgtctgcggatcttggaca atactgcctgcctgaagctcttgt ccagatggtcaaagggataca accactctccctttgcagaac ccgtttgtgggtccagca 214 ASMase . DControl eye - * g 03 —1. I l/R eye _5- . 2 e g 0.6 —I 0 i :1 i e 0.4 —«i 0 ; (I) i g ; l: 0.2 ‘i i 0 _L_ ASMase +/+ Figure 5.1. Retinal gene expression of ASMase in the HR retinopathy model. RT- PCR analysis of ASMase in I/R injured retinas isolated from ASM” mice 2 days following retinal ischemia-reperfusion injury. The relative amounts of mRNAs were calculated by using the comparative CT method. Cyclophilin was used as a control ail/(1 all resuIts were normalized to the abundance of cyclophilin mRNA. (n= 7 for ASMase group. P < 0.05 compared to ASMase“+ control eye). 215 NSMase 3 * [:1 Control eye 2-5 I IIR eye Transcript/ Cyclophilin ASMase +5“ ASMase "’ Figure 5.2. Retinal gene expression of NSMase in the [IR retinopathy model. RT- PCR analysis of NSMase in I/R injured retinas isolated from ASMase“+ and ASMase"" mice 2 days following retinal ischemia-reperfusion injury. The relative amounts of mRNAs were calculated by using the comparative CT method. Cyclophilin was used as a control and all results were normalized to the abundance of cyCIOphilin mRNA. (n= 7 for ASMaseW group, n= 3 for ASMase‘/' group. *P < 0.05 compared to ASMase“+ control eye, ASMase”+ I/R eye and ASMase"" control eye). 216 ICAM-l C 0.15 E E] Uninjured eye 2 . . g 0-1 Il/R Injured eye 0 E 'c 0.05 O U) C ('3 FL' 1- 0 _ ,- _ . ASMase I” ASMase"' B. 0.3 , VCAM-l E E Q. (_3 0 >5 0 b .9- b (D C 9 l—. ASMase 1" ASMase "' Figure 5.3. Retinal gene expression of cellular adhesion molecules in the HR retinopathy model. RT-PCR analysis of ICAM-1(A) and VCAM-l (B) in UK injured retinas (black bars) isolated from ASMase“+ and ASMase'/' mice 2 days following retinal ischemia-reperfusion injury. The relative amounts of mRNAs were calculated by using the comparative CT method. Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRNA. (n= 7-9 for ASMase“+ group, n= 5 for ASMase'/' group. *P < 0.05 compared to ASMase“ control eye and ASMase'/' control and I/R eye). 217 IL-lB TNFa .E 06 0.02 E * 3 0.015 1 g 0.4 1 I O : E 'z 0.2 8 1' c: (0 I: 0 ——~---— -- - —*—- ----—~—— --- ASMase +’+ ASMase"‘ ASMase +/+ ASMase 4’ C. VEGF .5 1 E o. 2 o > 0 0.5 :1 .9- '5 (D C re f: 0 ASMase 7“ ASMase "' Figure 5.4. Retinal gene expression of inflammatory/angiogenic molecules in the I/R retinopathy model. RT-PCR analysis of IL-lB (A), TNFa (B) and VEGF (C) in I/R injured retinas isolated from ASMase“ and ASMase“ mice 2 days following retinal ischemia-reperfusion injury. The relative amounts of mRNAs were calculated by using the comparative CT method. Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRNA. (n= 6-9 for ASMase“+ group, n= 5 for ASMase"" group. ‘P < 0.05 compared to ASMase” control eye and ASMase"' control and I/R eye). 218 ASMase i” I Uninjured eye I I l/R injured eye I ‘u-v weir, " ‘.‘ ."‘.1 . . --‘ A». ‘i ’. ‘. 'l g\..._' 1‘ _>.'4 ‘I ‘ ''r ‘l"- ‘ "t' “A '1“ ‘A.(:”‘ .‘ ‘ * Q . sex; Figure 5.5. Retinal capillary degeneration in retinal ischemia/reperfusion in [IR retinopathy mouse model. Acellular capilla occurrence (black arrows) in the retina isolated from (A) ASMase“+ and (B) ASMase" ' mice 7 days following retinal I/R injury. 219 ASMase 4' Uninjured eye l/R injured eye ‘~.)\l’ I . f Figure 5.5 (continued). Retinal capillary degeneration in retinal ischemia/reperfusion in [IR retinopathy mouse model. 220 _s 01 .L O 01 Acellular capillaries/mm2 0 “fin-“ __ __, ASMase ”L” ASMase "' [j Uninjured eye I IlR injured eye Figure 5.5 (continued). Retinal capillary degeneration in retinal ischemia/reperfusion in [IR retinopathy mouse model. (C) Quantification of the number of acellular capillaries fi'om five independent sets of mice, with a total of seven mice per ASMase” group and six mice per ASMase'I' group. At least eight fields of retina were counted in duplicates by two independent investigators; *P<0.05; red bar = 550nm, black bar = 110nm. 221 60 50 40 30 *' 20 - 10 Number of endothelial cell nuclei (neovascularization) - ASMase W .__J E! ASMase "' I Figure 5.6. Retinal neovascularization in oxygen-induced retinopathy in mice. Quantification of the endothelial cell nuclei representing abnormal retinal neovascularization in the ASMase” and ASMase'l' mouse retina. (n= 8 per ASMase” group and n= 6 per ASMase’/' group; 1IP