. tar—KL; . 5 .r «.3 r M .thfl 4. .m. a. .w W Q Q .Lo . . a... _ ”M; . '1'”- ..v aw. 1.... a. 2.. . I... . , a . .m . , .mmwqu mm? mm.» a . _ . LP“ $61.“. .. u..— 3. .4 2. arm/WVJQKHW. p... “Emma? r. a v .3 . . .~.. . t Ly, , . . fininnnzw r... . . has... . .1 —p . . u: . t .— ‘ ‘ . . U fixagfi? st...- Jvmt %. .2 ... r, . .2 .wM . ... “$3...“va ”fawn fiaghgwhuwwfiw . w... _ .1: a o x. .15... T x a. 2...", h. EL“? man. a}? afififimér ” n 4.35». a . m. . . m firfimfimfi . , , _. A. .m an”. .. m. .mxn... ,. 5......“ . km 7.“ r4. x... .12.... . :32.” . r. ._fi.....¥v 3:1}...541 - .I. 7. ‘ .35... wag“... . n .r "Mr. “tau 1. at. .. «Km. \5- a . Rd. , fl... Ru... 1.," Ii... “"35 924L939 _ .. .Q. . .\o$3€.. . .rw. rHESlS 2 Zoe] This is to certify that the dissertation entitled Studies elucidating the mechanisms of delta—9-THC -mediated protection from streptozotocin-induced diabetes presented by Xinguang Li has been accepted towards fulfillment of the requirements for Ph.D. Pharmacology & Toxicology degree in —Environmental Toxicology Z/Zé; Mm professor Date MM MS U is an Affirmative Action/Equal Opportunity Institution 0-12771 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 6/01 cycincmaeouopssms STUDIES ELUCIDATING THE MECHANISMS OF Ag-THC-MEDIATED PROTECTION FROM STREPTOZOTOClN-INDUCED DIABETES By Xinguang Li A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Pharmacology & Toxicology Institute for Environmental Toxicology 2000 ABSTRACT STUDIES ELUCIDATING THE MECHANISMS OF Ag-THC-MEDIATED PROTECTION FROM STREPTOZOTOClN-INDUCED DIABETES By Xinguang Li Ag-Tetrahydrocannabinol (Ag-THC) is a well-established immune suppressive agent, but has not been evaluated in models of immune-mediated diabetes. The overall objective of the present study was to investigate the protection by Ag-THC from the development of autoimmune diabetes. Multiple low dose streptozotocin (MLDSTZ)-induced diabetes in CD-1 mice has been utilized as an experimental model to study beta-cell pathogenesis in autoimmune diabetes. it was demonstrated in the current investigation that 11-day treatment with Ag-THC was able to attenuate the MLDSTZ-induced elevation in serum glucose and loss of pancreatic insulin in CD-1 mice. Additionally, MLDSTZ- induced insulitis and increases in IFN-y, TNF-oc and lL-12 mRNA expression were all reduced by co-administration of AQ-THC. In separate studies, Ag-THC given after completion of STZ treatment was found equally effective in protecting CD-1 mice from MLDSTZ-induced diabetes. Additionally, results from comparative studies showed that the BBC3F1 mice treated with MLDSTZ exhibited an absence of insulitis, but demonstrated moderate hyperglycemia and insulin loss. The less severe diabetic state in BGC3F1 mice was not altered by AQ-THC, suggesting that MLDSTZ treatment can initiate a diabetic state without an inflammatory response in pancreatic islets. Accordingly, these studies indicate that AQ-THC is capable of attenuating the severity of the autoimmune response in the experimental model of immune-mediated diabetes. The protection from MLDSTZ-induced hyperglycemia by Ag-THC treatment could also be attributed to a possible insulinotropic effect of the cannabinoid on CD-1 mice. Studies toward an understanding of the direct effect of A9-THC on pancreatic beta-cell function indicated that AQ-THC was able to stimulate insulin release from CD-1 mouse islets and rat-derived insulin- producing RINm5F cells. Closely associated with Ag-THC-stimulated insulin release was a calcium influx-induced elevation in intracellular calcium and an activation of CaM kinase II. Subsequent analysis of pancreatic islets and Rle5F cells identified mRNA expression for the cannabinoid receptor, CB1. Furthermore, the CB1 receptor antagonist, SR141716A was shown to block A9- THC-stimulated insulin release and elevation in intracellular calcium. These results indicate that Ag-THC-stimulated insulin release is mediated through an elevation of intracellular calcium through the opening of calcium channels via a CB1-dependent mechanism. In conclusion, the present study indicates that a suppression of cell-mediated immune response against pancreatic beta-cells and possibly a direct stimulatory effect on insulin release from beta-cells may be involved in AQ-THC-attenuated hyperglycemia produced by MLDSTZ treatment. To my parents, Chengxiao Li and Xiuqi Zhang To my husband, Wei Sun To my daughter, Lily for their love, their sacrifice and their support ACKNOWLEDGEMENTS I would like to give my deepest gratitude to my academic advisor, Dr. Lawrence Fischer for his masterful guidance, financial support and patience. The scientific discipline he provided allowed me to find the right direction for my graduate study and will greatly benefit my future career as well. I would also like to give my sincere appreciation to Dr. Norbert Kaminski. His excellent insight, his support and his encouragement really means very much throughout my graduate work. I felt particularly fortunate to have both him and Dr. Fischer as my research mentors. Many thanks go to my committee members Dr. Jack Harkema and Dr. Patricia Ganey for their shared expertise, their interest, and their encouragement. I want to especially thank our department chairperson, Dr. Kenneth Moore for his support and his help. The whole Kaminski lab and the Fischer lab, especially Robert Crawford, Robin Condie, Courtney Sulentic, Amy Herring, Barbara Faubert and Tony Jan, have been invaluable for their constructive suggestions. A special note of gratitude should go to Robyn Johnson, Kathy Campbell, and Amy Porter for their cheerful technical assistance. I would also like to thank my fellollow students Hyn Youn Cho for her friendship throughout the years. Finally, I would like to give my heartful appreciation to my husband, Wei Sun. Without his help, support and burden sharing, the degree and dissertation would have been beyond my reach. TABLE OF CONTENTS LIST OF TABLES ............................................................................................... IX LIST OF FIGURES .............................................................................................. X CHAPTER I INTRODUCTION ................................................................................................ XII I.1. Preface ................................................................................................ xiii |.2. Insulin and Insulin-producing cells ....................................................... xiii l.2.A. Morphology and physiology of the pancreatic islets ......................... xiii l.2.B. Functions of insulin—producing cells: insulin synthesis and insulin release ............................................................................................ xv I. 2. B. i. Insulin synthesis ................................................................... xv I. 2. B.‘ Ii. Insulin release and calcium-dependent signaling .............. xviii I. 3. Streptozotocin- induced diabetes ....................................................... xxiv I. 3. A. Diabetes: general background ....................................................... xxiv I.3.B. Streptozotocin: structure and biological effects .............................. xxvi I.3.C. Direct treptozotocin cytotoxicity ..................................................... xxix I.3.D. Multiple low dose streptozotocin-induced diabetes model ............. xxxii I.3.D.i. Genetic susceptibility ........................................................ xxxiii I.3.D.ii. Cell-mediated immune response ..................................... xxxv |.4. Cannabinoid compounds and Ag-THC ............................................. xxxix l.4.A. Cannabinoids: structure and biological effects .............................. xxxix I. 4. B. Cannabinoid receptors: 081 and C82 ............................................ inv I. 4. C. Intracellular signal transduction ...................................................... xlvii l ..4 D. Immune modulation by Ag-THC ........................................................... Ii I.5. Summary .............................................................................................. Iiv CHAPTER II EXAMINATION OF THE IMMUNOSUPPRESSIVE EFFECT OF A°-THC IN MLDSTZ-INDUCED DIABETES MODEL ........................................ LVI ”.1. Summary ............................................................................................ Ivii “.2. Introduction ........................................................................................ Iviii ".3. Materials and methods ........................................................................ lix II.3.A. Chemicals ........................................................................................ lix ”.33. Animals ............................................................................................. Ix II.3.C. Treatment ........................................................................................ 60 II.3.D. Quantitative competitive RT-PCR analysis. ..................................... 63 II.3.E. Histopathological examination. ........................................................ 65 II.3.F. Statistical analysis ............................................................................ 66 “.4. Results ................................................................................................ 66 vi II.4.A. Effects of 11 day AQ-THC treatment on MLDSTZ-induced hyperglycemia and loss of pancreatic insulin in CD-1 mice ............ 55 “.48. Effects of 11 day Ag-THC treatment on MLDSTZ-induced hyperglycemia and loss of pancreatic insulin in B603F1 mice ....... 60 II.4.C. Effects of Ag-THC treatment on MLDSTZ-induced insulitis in 00-1 and B6C3F1 mice .............................................................. 61 II.4.D. Effects of 11 day Ag-THC treatment on MLDSTZ-induced changes in mRNA expression of selected cytokines in CD-1 mice ............... 64 II.4.E. Effects of Ag-THC treatment post administration of MLDSTZ in CD-1 mice ................................................................................... 68 ”.5. Discussion .......................................................................................... 75 CHAPTER III CANNABINOID RECEPTOR CB1-MEDIATED INSULIN RELEASE FROM ISOLATED PANCREATIC ISLETS AND RINMSF CELLS ........... 84 ”L1. Summary ........................................................................................... 85 III.2. Introduction ........................................................................................ 86 "L3. Materials and methods ...................................................................... 88 |II.3.A. Materials ......................................................................................... 88 |II.3.B. Preparation of mouse pancreatic islets ........................................... 88 |II.3.C. Cells ................................................................................................ 89 ”L30. Measurement of insulin release ...................................................... 89 |II.3.E. Determination of cannabinoid receptor mRNA expression ............. 90 |II.3.F. Measurement of [Cam]i .................................................................... 92 |II.3.G. Statistical analysis .......................................................................... 93 III.4. Results ............................................................................................... 93 III.4.A. Stimulation of insulin release from mouse pancreatic islets and RINm5F cells by Ag-THC treatment ................................................. 93 ”L48. Ag-THC-induced increase in [Ca2*]i in RINm5F cells ....................... 93 III.4.C. Attenuation of A9-THC-induced increase in [Ca2*]i and insulin release by the calcium channel blocker verapamil .......................... 96 III.4.D. Thapsigargin and ryanodine exert no effect on the elevation in [Ca2+]i induced by Ag-THC ............................................................. 100 ”LS. III.4.E. Effects of CaM kinase II inhibitor KN93 and PKC inhibitor bisindolylmaleimide on the stimulation of insulin release by AQ-THC ..................................................................................... 100 III.4.F. Identification of cannabinoid receptor mRNA expression in isolated pancreatic islets and RINm5F cells ................................. 103 I ”46. Effects of cannabinoid receptor antagonists on Ag-THC- stimulated insulin release .............................................................. 106 III.4.H. Effects of cannabinoid receptor antagonists on Ag-THC- induced [Ca2*]i increase in RINm5F cells ...................................... 106 Discussion ....................................................................................... 1 13 vii CHAPTER IV OVERALL SUMMARY AND CONCLUSION .......................................... 120 BIBLIOGRAPHY ............................................................................................... 129 viii LIST OF TABLES Table ”-1. Histopathological evaluation of the protective effect of AQ-THC on MLDSTZ-induced insulitis in CD-1 mice ..................................... 67 LIST OF FIGURES Figure I-1. Diagram showing some important factors in glucose- stimulated insulin release ........................................................................ 9 Figure I-2. Chemical structure of streptozotocin and Vacor ......................... 17 Figure I-3. Chemical structure of Ag-THC and cannabinoid receptor antagonists ............................................................................. 29 Figure I-4. Scheme for the putative mechanism responsible for MLDSTZ-indcued autoimmune diabetes .................................................... 30 Figure "-1. Scheme of the Ag-THC and STZ treatment regimens ................... 50 Figure "-2. Effect of Ag-THC treatment on MLDSTZ-induced elevation of serum glucose in CD-1 mice and B6C3F1 mice .......................... 57 Figure "-3. Effect of Ag-THC treatment on pancreatic insulin in MLDSTZ-induced diabetes in CD-1 mice and B6C3F1 mice .......................... 59 Figure "-4. Representative photomicrograph of islets in pancreatic tissue .................................................................................. 63 Figure "-5. Representative photomicrograph of immunohistochemical staining of islets in pancreatic tissue obtained from CD-1 mice ...................... 66 Figure "-6. Effect of Ag-THC treatment on IFN-y RNA expression in MLDSTZ-induced diabetes in CD-1 mice ............................................... 70 Figure "-7. Quantitative analysis of cytokine steady state mRNA expression after A9-THC treatment in MLDSTZ-induced diabetes ................... 72 Figure "-8. Effect of Ag-THC post MLDSTZ-treatment on serum glucose and pancreatic insulin in MLDSTZ-induced diabetes in CD-1 mice .................. 74 Figure "-9. Scheme for the putative mechanism responsible for the protective effect of Ag-THC on the MLDSTZ-induced diabetes model ............................ 76 Figure III-1A. Effect of Ag-THC on insulin release from mouse pancreatic islets ......................................................................... 94 Figure III-1 B. Effect of Ag-THC on insulin release from RINm5F cells .............. 95 X Figure III-2. Effect of Ag-THC on [Ca2+], in RINm5F cells .............................. 97 Figure III-3A. Effect of calcium channel blocker verapamil on A9-THC-induced increase in [Ca‘P']i in RINm5F cells ................................. 98 Figure III-3B. Effect of calcium channel blocker verapamil on the stimulation of insulin release by Ag-THC in RINm5F cells .............................. 99 Figure III-4A. Effect of thapsigargin on Ag-THC-induced increase in [Ca2+], ............................................................................... 101 Figure III-4B. Effect of ryanodine on Ag-THC-induced increase in [Ca2*]i ....... 102 Figure III-5A. Effect of CaM kinase II inhibitor KN93 on the stimulation of insulin release by AQ-THC .................................................. 104 Figure III-SB. Effect of PKC inhibitor bisindolylmaleimide on the stimulation of insulin release by Ag-THC .................................................. 105 Figure Ill-6. mRNA expression of cannabinoid receptor in isolated pancreatic islets and RINm5F cells ............................................. 107 Figure III-7A. Effect of SR141716A on Ag-THC-stimulated insulin release from mouse pancreatic islets ...................................................... 108 Figure Ill-7B. Effect of SR141716A on Ag-THC-stimulated insulin release from RINm5F cells .......................................................... 109 Figure III-7C. Effect of SR144528 on AQ-THC-stimulated insulin release from RINm5F cells .......................................................... 110 Figure III-7D. Effect of cannabidiol on insulin release from RINm5F cells ...... 111 Figure III-8. Effects of SR141716A on Ag-THC-induced increase in [Ca2*]i in RINm5F cells .......................................................... 112 Figure III-9. CB1 receptor mediated Ag-THC-stimulated insulin release through calcium-dependent signaling pathways ........................................ 119 xi CHAPTER I INTRODUCTION l.1.Preface Current diabetes research is focused primarily on understanding the mechanisms of diabetes development and pursuing the potential for improved therapeutic treatments. Among those established therapeutic treatments, immune suppression has been previously demonstrated to be a potentially useful approach for immune-mediated diabetes. One well-known immunosuppressive agent currently under study is A9-tetrahydrocannabinol (Ag—THC). Ag-THC is the major psychoactive component of marijuana and alters a wide variety of biological effects including those associated with immune responses and release of insulin. The present study is undertaken to investigate the potential protective effect of Ag-THC on an immune-mediated diabetes model that employs multiple low dose streptozotocin (MLDSTZ) administration. This experimental model employed in the present investigation will further provide valuable insight into the mechanism by which certain environmental chemicals are capable of initiating immune-mediated diseases. To provide background and rationale for these studies, the remainder of the introduction section will first review the morphology and physiology of insulin-producing cells with particular emphasis on the regulation of insulin release. Furthermore, an overview of the literature relevant to STZ and the MLDSTZ-induced diabetes model will be presented. Finally, the therapeutic effects of Ag-THC and the related mechanisms will be introduced. l.2.lnsulin and Insulin-producing cells l.2.A.MorphoIogy and physiology of the pancreatic islets 2 Pancreatic islets are small spherical collections of endocrine cells scattered throughout the pancreas. Islets comprise approximately 1-2% of the mass of the pancreas. The remaining 98-99% of the pancreas is composed of the exocrine, ductal, connective and neural tissues (see review in (Bonner-Weir and Smith, 1994; Fischer, 1997)). The pancreatic Islets contain four major cell types, each of which is responsible for the synthesis and release of specific islet peptide hormones. Alpha-cells produce glucagon, beta-cells produce insulin, delta-cells produce somatostatin, and PP-cells produce pancreatic polypeptide. Generally, insulin- producing beta-cells are located in the center of the islets, and comprise approximately 70% of the islet mass. Alpha-cells, delta-cells and PP-cells are located around the islet periphery and make up approximately 30% of the islet mass. Individual islets have extensive networks of highly fenestrated capillaries allowing for rapid transfer of the islet hormones across the capillary endothelium into the circulation. Insulin, as one of the islet hormones, stimulates the uptake and utilization of glucose at various sites throughout the body, thereby lowering blood glucose concentration. Glucagon stimulates the mobilization and release of glucose, thereby raising blood glucose levels. Glucagon also stimulates the release of insulin, while insulin appears to inhibit the release of glucagon. Additionally, somatostatin inhibits the release of both glucagon and insulin. The net result of these tight regulations is that the concentration of blood glucose fluctuates only in a narrow range within normal physiological limits, despite fasting and food intake. Approximately 20% of the pancreatic blood flow is in the endocrine islets even though it represents only 1-2% of the total tissue mass. For larger islets, which obtain the majority of the islet blood flow, the arterial supply enters the islet at a discontinuity or break In the mantle cells. Capillaries radiate into the center of islet cells ending in collecting venules located at the periphery. As a result, the arterial blood containing a potential toxic substance may enter the islets and first contact insulin-producing beta-cells in the center of the islets. This blood flow pattern and the relatively high blood flow rate in the pancreatic islets may contribute to the high sensitivity of insulin-producing beta-cells to certain chemicals such as streptozotocin (STZ). As the blood moves from the islet core to the mantle, glucagon and somatostatin cells would be the next targets exposed to a potentially toxic chemical. This exposure and a possible change in normal insulin levels on insulin-producing cells due to chemical-induced effects could subsequently alter the normal release of glucagon and somatostatin. Therefore, an anatomical explanation for beta-cell selective toxicity is possible. I.2.B.Functions of insulin—producing cells: insulin synthesis and insulin release I.2.B.i.lnsulin synthesis The process from insulin genes to the formation of mature Insulin molecules in secretory granules is referred to as insulin synthesis (Steiner and Tager, 1979; Permutt.M.A, 1981; Permutt et al., 1984; Steiner et al., 1985; 4 Selden et al., 1987; Steiner, 1967). In general, the insulin gene is transcribed into preproinsulin mRNA (PPImRNA) by RNA polymerase II. The primary transcript of PPI is processed within the nucleus to form mature PPI mRNA through a post-transcriptional process, which involves an addition of a 7- methylguanosine residue at the 5' end, polyadenylation at the 3' end, and the splicing out of one or two introns. The mature PPImRNA leaves the nucleus, is bound to ribosomes and translated to form PPI. PPI has a very short half-life of approximately 1 min, indicating that it is rapidly processed to form proinsulin. After the pre portion of the proinsulin is removed from the nascent polypeptide, the resulting proinsulin molecule is sequestered into the membranous vesicles of rough endoplasmic reticulum (ER). Subsequently, the proinsulin molecules are transferred in small membrane-bound vesicles to the Golgi region. There, packaging of proinsulin into secretory granules is initiated. As the granules mature, C-peptide is cleaved from the proinsulin molecule to produce insulin by two types of proteolytic enzymes: a trypsin-Iike protease and a carboxypeptidase B-Iike-enzyme. The insulin molecule then crystallizes with zinc to form the dense core of the mature insulin secretory granules (Emdin et al., 1980). Afterwards, release of insulin from the secretory granules occurs by exocytosis and the insulin crystal is discharged to the extracellular space. It is generally accepted that there are multiple levels of control in the regulation of insulin synthesis. Glucose as the principle physiological regulator of insulin synthesis and release appears to modulate many steps of this process. It has been previously shown that glucose directly modulates PPImRNA levels in cultured mouse, rat and human islets ( Rafiq et al, 2000; Giddings et al., 1985; Hammonds et al., 1987; Jarrett et al., 1967). Low glucose concentrations (2.8- 3.3 mM) lead to decreases in the levels of PPImRNA, whereas high glucose concentrations (17-28 mM) allowed the maintenance of PPImRNA levels or increased PPImRNA levels after culture at low glucose (Permutt and Kipnis, 1972; Hammonds et al., 1987). The increased PPImRNA levels by high glucose are due to either alteration of insulin gene transcription and/or stabilization of PPImRNA. In addition, both initiation and elongation involved in the translational aspects of PPI synthesis are also enhanced by high glucose (Welsh et al., 1986). Furthermore, glucose—stimulation has been shown to alter proinsulin synthesis and conversion of insulin without changing the levels of PPImRNA (Itoh et al., 1978; Rafiq, 2000). The effects of other agents on insulin synthesis have also been documented. Nutritional substances (Welsh et al., 1986; Prentki et al., 1994), cyclic adenosine 3'5' monophosphate (CAMP) (Nielsen et al., 1985), dexamethasone (Welsh et al., 1988), and cholera toxin (Welsh et al., 1988) can stimulate insulin gene transcription in insulin-producing cells. On the other hand, cyproheptadine (Miller et al., 1993), STZ (90mg/kg) (Permutt et al., 1984), FK506 (Tamura et al., 1995), cytokines (e.g. IFN-y and TNF-a) (Sternesjo et al., 1995), and fasting (Giddings et al., 1991) have been shown to decrease insulin synthesis. The pancreatic beta-cell is the major site for insulin synthesis and insulin release in amounts sufficient to regulate blood glucose (Itoh et al., 1978; Giddings et al., 1985). Until recently, it was believed that insulin synthesis and 6 insulin release was limited to beta-cells of the pancreatic islets. However, it has now been demonstrated that extrapancreatic tissues such as human placenta, mouse and rat brain, and the yolk sac of the developing rat fetus also may synthesize insulin (Devaskar et al., 1993; Liu et al., 1985; Muglia and Locker, 1984). The physiological role of insulin synthesis in these non-pancreatic tissues is unclear at this time. I.2.B.ii.lnsulin release and calcium-dependent signaling Insulin stored in mature secretory granules is released by exocytosis, which is a multistage process involving transport of granules to the plasma membrane, their docking, priming and finally their fusion with the plasma membrane (Howell and Tyhurst, 1982). Once formed, granules enter a large cytoplasm granule storage pool from which only a few percent are secreted per hour. More recently formed granules are selected for earlier release, while older granules accumulate and, if not secreted, eventually fuse with Iysosomes which recycle the granule constituents. The above description is of the stimulated pathway of insulin release, which is required for attenuating high glucose levels and other nutrient stimulus (Prentki et al., 1997). Basal level insulin release occurs via a constitutive pathway that does not involve secretory granules. This pathway releases relatively small amounts of insulin that is insufficient for controlling rising glucose levels associated with food consumption or increased metabolic demands (Malaisse, 1972). Glucose is the major physiological stimulus for insulin release. Hyperglycemia stimulates the release of insulin, whereas hypoglycemia stimulates the release of glucagon (Shi et al., 1996). Insulin release by glucose stimulation is regulated by several critical components that are currently identified in pancreatic beta-cells (Fig.I-1) (Henquin J.C., 1994). Specifically, metabolism of glucose in the pancreatic beta-cells is initiated primarily by the rate-limiting enzyme glucokinase. This glucose metabolic signal results in an increase of the ATP/ADP ratio, which closes ATP-sensitive K+ channels (Km channels) in the plasma membrane. This closure decreases the K“ conductance, which allows the depolarization of the plasma membranes in the pancreatic beta- cells (Roenfeldt et al., 1992; Findlay and Dunne, 1985). For a long time, depolarization of the cell membrane was a property believed to be confined to a small group of highly sophisticated cells such as nerve and muscle cells in which the role and need of electrical signaling was obvious (Kostowski and Tarchalska, 1972; Cohen, 1972). However, during the 19608 and 1970s it became obvious that a number of endocrine cells share this capacity. They utilize changes in their membrane potential in order to transduce changes in their environment to acceleration of hormone secretion (Bernardis and Frohman, 1971; Phelps and Sawyer, 1977). The pancreatic beta-cell is one of the examples. Dean and Matthews provided the first evidence for glucose-stimulated membrane depolarization in the insulin-producing beta-cells (Dean and Matthews, 1968; Dean and Matthews, 1970). When this depolarization is large enough, voltage- dependent calcium channels become activated. The initial depolarization up to the threshold potential causes the opening of a few voltage-dependent calcium Glucagon Protein [mp kinase A “— IX 91 GLP-l GIP Protein fl kinase E \ DAG Insulin Secretion Figure I-1. Schematic representation of the major mechanisms by which stimulants and inhibitors regulate insulin release: -: inhibition; +; stimulation; Ach: acetylcholine; DAG: diacylglycerol; CaM kinase: calcium/calmodulin dependent kinase; IP3: inositol 1,4,5-triphosphate; PIP2: phosphotidylinositol 4,5- biphosphate; GLP-1: glucagonlike peptide; 6le gastric inhibitory polypeptide (Modified from reference - Henquin, 1994). channels, which in turn causes a bigger depolarization and the opening of additional calcium channels. The calcium influx via opening of calcium channels produces a transient elevation of the intracellular Ca2+ concentration ([Ca2*],) followed by a series of reactions including protein phosphorylation. These events culminate in the exocytosis of the insulin-containing granules (Ashcroft and Hughes,1990) In addition to glucose, a variety of stimulants such as hormones, neuropeptides, and sulfonylurea drugs can also stimulate insulin release from pancreatic beta-cells (Figl-1) (Prentki et al., 1997; Neye and Verspohl, 1998). In certain stimulus-secretion coupling, a calcium-independent mechanism (e.g. ATP-dependent) has been proposed and reported as an alternative signaling pathway (Sakuma et al., 1995; Lang et al., 1998). However, the [Caz*]i plays a fundamental role in most secretagogue-induced release of insulin (Zawalich et al., 1998; Ashcroft & Rorsman, 1989; Wollheim and Pozzan, 1984). Metabolism of glucose and sulfonylurea drugs stimulate release of insulin by producing an Increase in [Ca2"]i through a calcium influx into pancreatic beta-cells, via voltage- dependent calcium channels (Porksen et al., 1996; Seri et al., 2000). In addition, stimulants like carbachol or 12-O-tetradecanoylphorboI-13-acetate (TPA), may mobilize Ca2+ from intracellular calcium stores, resulting in an increase in the [Ca2+]i and insulin exocytosis (Zawalich et al., 1998; Tang et al., 1995). Current evidence suggests that release of Ca” from intracellular compartments in pancreatic beta-cells can be triggered by activation of various intracellular Ca2+ release channels (Ilkova, 1994). Insulin-producing cells may contain either or 10 both types of intracellular Ca2+ release channels: 1) inositol 1,4,5-trisphosphate (IP3) receptor/ Ca2+ release channel; and 2) ryanodine receptor/CaZ*-induced Ca2+ release channel (Lange and Brandt, 1993; Takasawa et al., 1998; Holz et al., 1999). To determine the involvement of calcium in a particular cell function, various Ca2+ channel blockers or inhibitors have been commonly utilized. Garcia et al. employed verapamil in reducing the movement of calcium into the cells from the extracellular matrix and indicated a calcium influx involved in the adrenoceptor-stimulated insulin release (Garcia et al., 1992; Ohta et al., 1993). Inhibitors for ER Ca2*-ATPase such as thapsigargin (Aizawa et al., 1995), or inhibitors acting at ryanodine receptor-operated channels for Ca2*-induced Ca2+ release (Holz et al., 1999) have also been commonly employed to study the regulation of Caz+ transport in beta-cells. The mechanisms by which most secretagogues stimulate insulin release involve a calcium-dependent signaling pathway. However, the precise regulatory mechanism by which a sufficient increase in [Ca2+], is translated into the release of insulin is still largely unknown. Current reports implicated that calcium should perhaps be regarded as an initiator rather than a determinant of exocytosis (Easom, 1999). It has been suggested that the amplitude of the exocytotic responses depended to a greater extent on the activity of protein kinases and phosphatases than the actual [Ca2*]i (Braiman et al., 1999; Miura et al., 1998; Cui et al., 1998). For example, agents which increase intracellular cAMP levels, such as glucagon and glucagon-like peptide-1 (GLP-1), were able to potentiate glucose-stimulated insulin secretion almost 10-fold while modestly calcium influx 11 and [Ca2+],, suggesting a protein kinase A (PKA)«dependent mechanism (Gromada et al., 1997). However, no stimulation of exocytosis was observed in the absence of glucose, indicating that glucose metabolism was required for the PKA—dependent phosphorylation. In addition, calcium-dependent exocytosis was also enhanced by agents which activated protein kinase C (PKC), such as acetylcholine (Hughes et al., 1990) and the phorbol ester 4-8-phorbol-12-B- myristate-13-a -acetate (PMA) (Howell et al., 1990). In general, conditions which promote protein phosphorylation lead to enhancement of secretion. Conversely one would expect that agents which produce the activation of protein phosphatases inhibit exocytosis. Indeed, this seems to be the mechanism by which the inhibitory hormones and neurotransmitters somatostatin, galanin and adrenaline suppress glucose-stimulated insulin secretion. The action of these compounds is mediated by activation of an inhibitory (pertussis toxin-sensitive) G-protein and culminates in the activation of the protein phosphatase calcineurin. Accordingly, numerous Ca2*—dependent protein kinases and phosphatases have been demonstrated within secretory cells, and many of these are implicated in the secretory process. A particular important member in the calcium-dependent protein kinase family is the group of Ca2*/CaM-dependent kinases (CaM kinases). The prototype of this group is smooth muscle myosin light-chain kinase (MLCK), which phosphorylates myosin regulatory light chain in a CaZVCaM-dependent manner resulting in activation of actomyosin ATPase required for muscle fiber contraction (Walsh et al., 1982; Kilhoffer et al., 1992). A CaM kinase distinct from 12 MLCK was identified in a rabbit brain extract (Endo and Hidaka, 1980) and many CaM kinases have subsequently been discovered. They are classified into two groups based on their ability to phosphorylate the endogenous protein target: 1) multifunctional enzymes: CaM kinase IN, II, and IV; and 2) specific enzymes: MLCK and CaM kinase III (Kishi et al., 1998; Sobieszek, 1994; Wenham et al., 1994; Nairn and Picciotto, 1994). Of these kinases, several have been identified in pancreatic beta-cells including MLCK, CaMII and CaMKIlI (Wenham et al., 1994; Niki et al., 1993; Mohlig et al., 1997). It has been demonstrated that the loss of secretory responsiveness of islets to Ca” is accompanied by reductions in calcium-dependent phosphorylation of endogenous islet substrates for CaM kinase II (Norling et al., 1994). This suggests an important role for CaM kinase II in beta-cell stimulus-secretion coupling. The inhibitors of CaM kinase II, such as KN93 have been useful in studying the activation mechanism of CaM kinase II. Treatment with KN93 (up to 10 pM) suppressed glucose-induced insulin release in isolated rat pancreatic islets in a dose-dependent manner without affecting its basal secretion (Wenham et al., 1994). By contrast, insulin release stimulated by TPA, a direct activator of PKC, was not affected by KN93. Taken together, Ca2+ plays a critical role in insulin release and CaM kinase u may be involved in the Ca2*-mediated insulin release. I.3.STZ-induced diabetes I.3.A.Diabetes: general background Diabetes mellitus represents a complex variety of disorders characterized 13 by the inability to regulate blood glucose levels within normal physiological limits (Walsh et al., 1982). At the molecular level, diabetes can occur from impaired insulin secretion and/or insulin action. On this basis, diabetes has been classified into type I (insulin-dependent) and type II (non-insulin-dependent) diabetes mellitus (see review in Joslin’Diabetes, Krall et al., 1994). Type I diabetes often occurs at a young age, while type II diabetes occurs later in life. Since type II diabetes results primarily from impaired insulin-induced signaling rather than insulin secretion from the beta-cell, it is not the focus of the present study. Type I diabetes results from an almost complete destruction of pancreatic beta-cells with maintenance of the alpha-cells and delta-cells within islets of Langerhans (Gepts, 1965). In parallel with dramatic developments in genetics and immunology during recent years, it has become clear that type I diabetes develops as a consequence of the selective destruction of pancreatic insulin- producing cells by autoimmune mechanisms. Although the etiology responsible for type I autoimmune-based diabetes has not yet been definitively elucidated; it is currently believed that some environmental factors are one of the elements that contribute to the disease and may trigger its onset. These factors could include mycobacteria, viral infections, stress, sex hormones and exposure to xenobiotics such as certain drugs, food constituents, and environmental toxic chemicals (Cahill and McDevitt, 1981; Craighead, 1978; Yoon, 1990). The first case was reported from a clinical investigation involving individuals who ingested rodenticide Vacor and later became diabetic (Karam et al., 1980; Prosser and Karam, 1978). Other known beta-cell toxicants include the marine antifoulant 14 triphenyltin fluoride (Manabe and Wada, 1981), antiserotonergic drug cyproheptadine (Fischer et al., 1973), the experimental diabetogenic agent alloxan (Veleminsky et al., 1970), and the nitrosourea compound STZ (Rakieten et al., 1963). Among these compounds, the rodenticide Vacor and STZ represent the group of N-nitrosamines (Fig.I-2). N-nitrosamines are present in a variety of food constituents and also can be formed in the body after ingestion of conventional food such as dried bacon, spinach, and nitrate/nitrite-rich drinking water (Brown, 1999; Scanlan, 1983). Contamination of nitrosamines has also been found in rubber nipples for babies’ bottle and many other rubber product (Havery and Fazio, 1982; Fajen et al., 1979). Accordingly, these compounds pose a potential threat to humans either through trace contaminants in the environment or through exposure to formation of these agents in the body, and therefore have generated considerable public concern because of their potential to cause diabetes and their known ability to produce cancer. The better understanding of their biological effects and the underlying mechanisms may contribute to both environmental toxicology studies as well as clinical diabetes research. I.3.B.Streptozotocin: structure and biological effects STZ was first discovered in 1959 by the UpJohn company (Wiggans et al., 1959). It was used as a broad-spectrum antibiotic as well as an antitumor agent for malignant beta-cell tumors. After its diabetogenic effect was found in rats and dogs in 1963 (Rakieten et al., 1963), STZ has been widely used to produce an 15 animal model of experimental diabetes. The effective dose of STZ in inducing diabetes ranges from 50 to 200 mg/kg depending upon the species, such as rats, mice, dogs, Chinese hamsters, monkeys, miniature pigs, and rabbits (Lazarus and Shapiro, 1972). Certain species such as guinea pigs and Syrian hamsters are much more sensitive to STZ than to alloxan, another commonly used diabetogenic chemical (Veleminsky et al., 1970). The structure of STZ has been determined to be the nitrosamide methylnitrosourea linked to the C-2 position of D-glucose and exists as either the a- or 8- anomers in the pyranose form (Fig.l-2). STZ is very unstable in solution even at acid pH, and should be administered promptly after being dissolved in citrate buffer at pH 4.5. The in vivo life span of STZ is less than 15 min (Aganival, 1980) It has been suggested that the plasma membrane glucose transporter is involved in the entry of STZ into pancreatic beta-cells. The a-anomer of STZ, similar to the a-anomer of glucose, showed greater effect on insulin secretion (Malaisse, 1991; Rossini et al., 1977). In addition, cells that have lost their responsiveness to glucose (e.g. RlNr cells) also have lost their sensitivity to STZ toxicity (LeDoux and Wilson, 1984). It has also been demonstrated that analogues of STZ whose glucose moieties were replaced by manonose, galactose, or alpha-O-methylglucose were nondiabetogenic (Bannister, 1972; Kawada et al., 1986). Moreover, studies with 1“C-STZ showed that considerably more STZ was taken up by the beta-cell than nitrosamide methylnitrosourea (MNU) which does not have the glucose moiety (Wilson et al., 1988). These 16 0 N 250 mg/dL when tested by Uristix strips (Bayer Co., Elkhart, IN)] starting on Day 9 (data not shown). The serum glucose (Fig.ll-2) and pancreatic insulin (Fig.ll-3) for the CD-1 mice in each of the different treatment groups was determined on Day 11 unless indicated otherwise. Serum glucose in the STZ (40 mg/kg/day) treatment group was observed to be in excess of 400 mg/dL, indicating severe diabetes. Mice receiving STZ in combination with Ag-THC (150 mg/kg/day) given on Day 1 through Day 11 exhibited significantly lower blood glucose concentrations than the STZ treatment group, i.e. 312.1:156 mg/dL (STZ+11dTHC) versus 447.0i9.7 mg/dL (STZ). No severe hyperglycemia or positive urine glucose was observed in the STZ+11dTHC treatment group. However, compared to the VH 55 FIGURE "-2. Effect of Ag-THC treatment on MLDSTZ-induced elevation of serum glucose in CD-1 mice and BSC3F1 mice. Untreated naive (NA) mice were utilized as a control for the vehicle (VH) treatment group. The streptozotocin (STZ) treatment group received 5 consecutive 40mg/kg i.p. injections on Days 1-5. The 11dTHC treatment group received 11 consecutive doses of Ag-THC (150 mg/kg) by oral gavage on Days 1-11. The STZ+11dTHC treatment group received 5 consecutive STZ (40mg/kg) i.p. injections on Days 1-5 and 11 consecutive doses of Ag-THC (150 mg/kg) by oral gavage on Days 1-11. Serum glucose was measured on Day 11 and Day 17 (see insert). The results are expressed as the mean i SE, n=15. A significant difference of p<0.05 is indicated where “a” denotes mean values that are different compared to the VH group for CD- 1 mice and “b” denotes mean values that are different compared to the STZ group for CD-1 mice; for B6C3F1 mice, “a' ” denotes mean values that are different compared to the VH group. 56 (1p/6w)asoonle POOIB STZ STZ 11dTHC VH 11dTHC Treatment FIGURE 3. Effect of Ag-THC treatment on pancreatic insulin in MLDSTZ- induced diabetes in CD-1 mice and BGC3F1 mice. The treatments were performed as described in Figure 2 legend. Pancreatic insulin was measured on the samples taken on Day 11. The results are expressed as the mean t SE, N=15. A significant difference of p<0.05 is indicated where “a” denotes mean values that are different compared to the VH group for CD-1 mice and “b” denotes mean values that are different compared to the STZ group for CD-1 mice; for 86C3F1 mice, “a' ”denotes mean values that are different compared to the VH group. 58 / / B6C3F1 .0 (U O I 11dT .0 I > S, .Q 300 - I ' I ‘ I fl I 7 I f I o o o o o 0 Lo 0 to o In N N ‘— ‘— (enssg; Bwlfiu) uunsul oneeioued Treatment 11dTHC group (192.0i27.2 mg/dL), STZ+11dTHC mice attained a mild hyperglycemic state with serum glucose concentrations above 250 mg/dl. No significant difference in serum glucose was observed among the NA, VH and 11dTHC treatment groups. Additional measurements were obtained on Day 17, 6 days after Ag-THC administration was terminated on Day 11, to determine whether the protective effect of Ag-THC was persistent (data shown in Fig.ll-2 insert). The results indicated that serum glucose, in contrast to results obtained on Day 11 was severely elevated on Day 17 in the STZ+11dTHC treatment group and equal to that in the STZ given alone treatment group. Pancreatic insulin in MLDSTZ-treated and control animals measured on Day 11 indicated that the treatment alone lowered pancreatic insulin content to less than 25% of control. STZ+11dTHC treated mice exhibited higher pancreatic insulin content than mice in the STZ only treatment group, i.e. 95.3:154 ng/mg pancreatic tissue (STZ+11dTHC) versus 35.1:85 ng/mg pancreas tissue (STZ) (Fig.ll-3). No significant difference in pancreatic insulin was observed among the NA, VH and 11dTHC treatment groups. |I.4.B.Effects of 11 day A°-THC treatment on MLDSTZ-induced hyperglycemia and loss of pancreatic insulin in B6C3F1 mice Identical studies to those described above for CD-1 mice were performed using B6C3F1 mice. Mice in the STZ treatment group did not show signs of polydipsia, polyuria, and positive urine glucose on Day 11 (data not shown), but 60 were mildly hyperglycemic when compared to the VH group (Fig.ll-2). Mice in the STZ+11dTHC treatment group showed no statistically significant differences in serum glucose from mice in STZ group. Similar results were observed when pancreatic insulin content was used as an index of STZ-induced destruction of pancreatic beta-cells (Fig.lI-3). Pancreatic insulin in the STZ group was decreased from vehicle control, i.e. 49.5i12.1 ng/mg pancreatic tissue (STZ) versus 189.4:10] ng/mg pancreatic tissue (VH). Mice in the STZ+11dTHC treatment group showed no statistically significant differences in insulin content from mice in the STZ treatment group. ll.4.C.Effects of A’-THC treatment on MLDSTZ-induced insulitis in CD- 1 and BGC3F1 mice Histopathological evaluation of pancreatic tissue was performed to examine the development of insulitis. The evaluation of tissues from CD-1 mice (Fig. “-4) demonstrated that the pancreatic islets of mice in the NA, VH, and 11dTHC-treatment groups were normal, i.e. few mononuclear cells present in the islets. In addition, Ag-THC treatment significantly attenuated the insulitis induced by MLDSTZ as evidenced by significantly fewer numbers of infiltrating lymphocytic cells. Lymphocytic cells, when present were primarily localized around the periphery of the islets as a result of the MLDSTZ + 11THC treatment. Compared to the mice in the STZ treatment group, disruption of islet cytoarchitecture and the presence of occasional necrotic cells within the islets were less pronounced in the STZ+11dTHC treatment group. Morphologically, the 61 FIGURE "-4. Representative photomicrographs of islets in pancreatic tissue obtained on Day 11: (A) VH/CD-1 mice; (B)11dTHC/CD-1mice; (C) STZ/CD-1 mice; (D) STZ+11dTHC/CD-1 mice; (E) VH/BGC3F1 mice; (F) STZ/BBC3F1 mice. For histopathology examination, pancreatic tissue was placed in 10% buffered formalin at sacrifice, and tissue sections were stained with H&E. (CD-1 mice: 100X ; 86C3F1 mice: 240x ). 62 63 STZ+6dTHC treatment group was similar to the STZ+11dTHC treatment group, i.e. a less pronounced degree of insulitis as compared to the STZ treatment group (data not shown). A semi-quantitative evaluation of the histopathology results is provided in Table 1. It indicates that the STZ+11dTHC group showed a moderate degree of insulitis as compared to the STZ group. Histopathology of tissues from BBC3F1 mice in contrast to results from CD-1 mice indicated that MLDSTZ treatment showed moderate to no insulitis when compared to vehicle controls (Fig.ll-4). Due to this observation, further studies in which cytokine levels in MLDSTZ-treated animals were measured employed CD-1 mice that showed severe insulitis on Day 11. The results in Figure ”-5 illustrate that the majority of inflammatory cells infiltrating the pancreatic islets are T-lymphocytes as measured by immunohistochemical staining. Pancreatic tissue sections were stained with anti-CD3+ antibody. The islets from the VH control group showed no CD3+ positive cells. Conversely, the pancreatic islets from STZ-treated mice showed marked infiltration of inflammatory cells, the majority of which were CD3+ positive cells. Compared to the STZ group, the STZ+11dTHC treatment group exhibited markedly fewer CD3" positive cells within the islets, although some were observed on the periphery of islets. ll.4.D.Effects of 11 day A°-THC treatment on MLDSTZ-induced changes in mRNA expression of selected cytokines in CD-1 mice To determine the profile of cytokines produced in pancreatic tissue during MLDSTZ-induced insulitis, RT-PCR was employed to evaluate total pancreatic 64 FIGURE "-5. Representative photomicrographs of immuno-histochemical staining of islets in pancreatic tissue obtained from CD-1 mice on Day 11, 100x. Pancreatic tissue was fixed with 10% buffered formalin. Tissue sections shown in (A)VH, (C)STZ, and (D)STZ+11dTHC were incubated with anti-CD3” and tissue section shown in (B)STZ was incubated with control serum. 65 66 Hislo indm MC (n Thu we fol TABLEI Histopathological evaluation of the protective effect of Ag-THC on MLDSTZ- induced insulitis in CD-1 mice. (Values indicate numbers of mice) TREATMENT GROUP ZESUIZEIEIEE NA VH 1 1dTHC STZ STZ+1 1dTHC NONE 14 15 15 0 3 MODERATE 1 O O 3 1O SEVERE O O 0 12 2 The results are compiled from three separate studies (N=15). Each pancreas was prepared for light microscopy then examined and placed into one of the following categories: a . . . . ' None: no Inflammation, normal Islet architecture; Moderate-insulitis: some lymphocytes infiltration surrounding but little infiltration into the islets Severe insulitis: massive lymphocytes infiltration into the islets with islet destruction. 67 RN. ST} ex; W9 an IIII Ire RNA in treated and control CD-1 mice. Qualitaive RT-PCR analysis showed that STZ induced significant increases in lL-12, lFN-y and TNF-a in mRNA expression on Day 11, as compared to the VH group; whereas, lL-2 and lL-4 were not detectable by RT-PCR analysis (data not shown). Quantitative RT-PCR analysis was performedto analyze selected cytokines. The time course of lFN-y mRNA expression was determined to examine the onset of insulitis. A significant increase in lFN-y steady state mRNA expression was observed in the STZ treatment group as compared to vehicle controls on and after Day 9 (Fig.ll-6). On Day 11, A9-THC treatment reduced the STZ-induced IFN-y (Fig.ll-7a), lL-12 (Fig. ll-7b) and TNF-a (Fig. Il-7c) steady state mRNA expression when 'compared to the STZ group. No significant difference in the magnitude of mRNA expression was observed for any of the measured cytokines among the NA, VH and 11dTHC groups. ll.4.E.Effects of A°-THC treatment post administration of MLDSTZ in CD-1 mice An abbreviated and delayed A9-THC treatment schedule was employed in which mice received 6 consecutive daily treatments with A9-THC (150 mg/kg/day) beginning on Day 6, one day after termination of the STZ treatment. A protective effect was observed similar to that produced by 11-day A9-THC treatment. Serum glucose concentrations in the STZ+6dTHC group were 286.5:74 mg/dL versus 447.6:10.1 mg/dL in the STZ treatment group (Fig.ll-8). Similarly, pancreatic 68 FIGURE "-6. Effect of AQ-THC treatment on lFN-y RNA expression in MLDSTZ-induced diabetes in CD-1 mice. The treatments were performed as described in Figure 2 legend. RT-PCR analysis for lFN-y was performed on the total pancreatic RNA isolated from the tissue samples taken from mice on the days indicated above. The results are expressed as the mean i SE, N=3; A significant difference of p<0.05 is indicated where “a" denotes mean values that are significantly different compared to the VH group. 69 \s D IIIIIIII O: O :3 O O O O O O O H) O O ‘— 0) O N O m ‘— (O OOOOOOOOOOO VVVVVV FIGURE "-7. Quantitative analysis of cytokine steady state mRNA expression after A9-THC treatment in MLDSTZ-induced diabetes. The treatments were performed as described in Figure 2 legend. RT—PCR analysis for lFN-y (a), lL-12 (b) and TNF-a (c) was performed on the total pancreatic RNA isolated from the tissue samples taken on Day 11. “ND” indicated |L-12 mRNA of NA, VH and 11dTHC groups were below the level of quantitative measurements. The results are expressed as the mean 1 SE, N=15; A significant difference of p<0.05 is indicated where “a” denotes mean values that are significantly different compared to the VH group and “b” denotes values that are significantly different compared to the STZ only group. 71 mRNA (mplecular/100 ng total RNA) 100000~ 80000- 60000- 40000- 20000- 0 - 1000 I 800 _ 600 — 400 q 200 — A (lFN-y) ND ND ND 0 800000- 600000- 400000- 200000- c (TNF-oc) a NA VH 1 1dTHC Treatment 72 STZ STZ+11dTHC FIGURE "-8. Effect of Ag-THC post MLDSTZ-treatment on serum glucose and pancreatic insulin in MLDSTZ-induced diabetes in CD-1 mice. Untreated naive (NA) mice were utilized as a control for the vehicle (VH) treatment group which received 0.2 ml citrate buffer, i.p., for 5 consecutive days and 0.2 ml corn oil, by oral gavage, for 6 consecutive days. The STZ treatment group received 5 consecutive 40mg/kg i.p. injections on Days 1- 5. The 6dTHC treatment group received 6 consecutive dose of Ag-THC (150 mg/kg) by oral gavage on Days 6-11. The STZ+6dTHC treatment group received 5 consecutive STZ (40mg/kg) i.p. injections on Days 1-5 and 6 consecutive dose of AQ-THC (150 mg/kg) by oral gavage on Days 6- 11. Serum glucose and pancreatic insulin were measured on Day 11. The results are expressed as the mean t SE, n=15. A significant difference of p<0.05 is indicated where “a” denotes mean values that are different compared to the VH group and “b” denotes mean values that are different compared to the STZ group. 73 m (anssg; Bun/Bu) uunsu' OnBSJOUBd O O O O O 8 8 O O O m N J ‘ I STZ 9. to \V ‘° STZ .o .o 5dTHC I > g o. \\ .Q uuuuuuuuuu - (1p/BUJ) 3803n|9 p00|8 4. Treatment insulin content in the STZ+6dTHC group was 106.2:212 ng/mg pancreatic tissue, which was significantly different from 35.1i3.5 ng/mg pancreatic tissue in STZ group (Fig.ll-8). ll.5.Discussion The results presented in the current study indicate Ag-THC can exert a transient attenuation from MLDSTZ-induced autoimmune diabetes. Both MLDSTZ-induced elevation of serum glucose and loss of pancreatic insulin are significantly diminished by Ag-THC co-treatment in CD-1 mice (Fig.lI-2, Fig.ll-3). MLDSTZ-induced insulitis is also significantly attenuated as evidenced by decreases in CD3 inflammatory cells in the pancreatic islets and in mRNA expression for IL-12, IFN-y, and TNF-a (summarized in Fig.ll-9). The protective effect of Ag-THC in this autoimmune model of diabetes is similar to previous results from studies in which treatments with Ag-THC, AB-THC and the synthetic cannabinoid compound, HU-211, were reported to significantly reduce the incidence and severity of experimental autoimmune encephalomyelitis in rats (Wirguin et al., 1994; Achiron et al., 2000; Lyman et al., 1989). These results indicate that cannabinoid compounds are able to diminish the severity of autoimmune responses in experimental models of autoimmune diseases. The inhibition of T-lymphocyte activation by cannabinoid compounds has been extensively demonstrated (Kaminski, 1996; Schatz et al., 1992; Condie et al., 1996; Klein et al., 1998). Based on previous findings, the protective effect of 75 mwcoamwm wczEE_ “5:325-th UBEEE 01.7% .8355 9865 $95 858 .685 38% 8035-584: 2. .5 05% .0 Soto 93020.5 9: .8 636589 EwEmcooE 3:33 9: .8 95:8 .m... 959“. 76 Ag-THC on MLDSTZ-induced insulitis may involve two possible mechanisms: either through suppressing T-lymphocyte infiltration into the pancreatic islets and/or by directly inhibiting T lymphocyte and macrophage function. Consistent with either mechanism, a marked decrease in inflammatory cells, including CD3 cells was apparent in mice treated with both Ag-THC and STZ as compared to those treated with STZ only. In addition, the reduction of lFN-y mRNA expression is consistent with the attenuation of MLDSTZ-induced diabetes by Ag-THC through inhibition of a TH1-mediated immune response. Based on the profile of lymphocyte cytokine production, T helper cells are divided into at least two distinct subsets, TH1 cells and TH2 cells. IL-12 produced by macrophages can stimulate IL-2 and lFN-y secretion that helps to initiate cell-mediated immunity. Most results from studies using animal models of spontaneous autoimmune diabetes suggest a dominant activation of TH1 over TH2 cells as being a key determinant in the pathogenesis of autoimmune diabetes (Maclaren and Alkinson, 1997; Rabinovitch, 1994). In the present study, total pancreatic mRNA was analyzed for the expression of TH1-associated cytokines including lL-2, IFN- y, lL-12 and the TH 2 cytokine, lL-4 on Day 11, six days after the last STZ treatment. The TH1 cytokines, specifically lFN-y, IL-12 and the proinflammatory cytokine, TNF-a, showed lower steady state mRNA expression associated with AQ-THC co-treatment as compared to the STZ only treatment group indicating a reduction in inflammatory response (Fig.ll-7). Expression of |L-2 could not be detected at the peak of MLDSTZ-induced insulitis in any of the experimental 77 treatment groups. The presence of TH1 cytokines such as IFN-y, IL-12 on Day 11 in the STZ-treated animals suggested that lL-2 maybe expressed prior to Day 11 as suggested by the studies of Herold et al. (Herold et al., 1996). The lack of detection in the limited sampling protocol used in this study does not permit speculation on the possible role of |L-2 and IL-4 in the MLDSTZ-induced diabetes model. The protective effect of Ag-THC on MLDSTZ induced diabetes could also be accomplished by acting at target sites other than the immune system. Cannabinoid compounds produce a number of biological effects on different cell types and organs systems. Therefore, alternative explanations for Ag-THC- mediated protection from MLDSTZ-induced diabetes were considered. The protective effect of Ag-THC could be mediated by several different putative mechanisms, such as: 1. inhibition of STZ uptake by beta-cells therefore blocking the direct cytotoxic action of STZ; 2. direct stimulation of glucose metabolism (Sanchez et al., 1998) resulting in reduced hyperglycemia; 3. direct effects on pancreatic beta-cell functions including increased insulin production and secretion as previously suggested (Laychock et al., 1986). One or more of these AQ-THC-mediated effects may account for the protective mechanism in the MLDSTZ model; however, several of these possibilities appear unlikely based on observations made in the present investigation and previously by others. For example, the AQ-THC treatment was also protective even when Ag-THC treatment was initiated 24 hr after administration of the last dose of STZ (Fig.ll-8). Due to 78 the very short half-life of STZ (approximately 15 min), STZ would have been cleared from the bloodstream and any direct STZ beta-cell toxicity would have been initiated (Like and Rossini, 1976). Thus interference by Ag-THC with STZ uptake or its initial action on the beta-cell appears unlikely. Furthermore, the notion that AQ-THC increases glucose metabolism to reduce hyperglycemia also seems to be contrary to our findings because Ag-THC treatment alone did not alter serum glucose and pancreatic insulin. The possibility that Ag-THC directly affects pancreatic beta-cell function(s) appears to be a plausible explanation for protection from hyperglycemia because there is evidence that insulin secretion can be stimulated by this cannabinoid. A study conducted by Laychock and colleagues showed that A9-THC stimulated the basal release of insulin and also potentiated glucose-stimulated insulin release in isolated rat pancreatic islets (Laychock et al., 1986). These actions would reduce hyperglycemia and may attenuate the loss of pancreatic insulin in the MLDSTZ + THC treated mice utilized in the experiments reported here. Based on this limited information, it is possible that immunosuppression alone may not account for the ability of Ag-THC to attenuate MLDSTZ-induced diabetes in CD-1 mice. Although Ag-THC attenuated MLDSTZ-induced diabetes in CD-1 mice, complete and permanent protection was not achieved. The lack of complete protection may be attributed to several factors. One possible explanation may be STZ produces irreversible cytotoxic damage to beta-cells that is insensitive to modulation by Ag-THC. To date, there has been considerable speculation 79 regarding the role of the immune response in the MLDSTZ model and whether or not damage to the beta-cells by the leukocytes is sufficient in itself to produce cell death (Beattie et al., 1980). It has been shown that STZ alone can directly Induce the lethal alkylation of cellular components and also can cause some nonlethal alterations in pancreatic beta-cell function (Sarvetnick et al., 1990; Maclaren and Alkinson, 1997). These alterations may include conformational changes in . membrane proteins associated with glucose transport and metabolism. These or other alterations may further be recognized as “non-self” bythe immune system and thus contribute to the immune-based beta-cell damage. Therefore, a suppression of either the direct cytotoxic effect by STZ on beta-cells or the subsequent inhibition of the immune response by A9-THC may only result in partial protection. The partial protective effect by Ag-THC treatment on MLDSTZ-induced diabetes in the present study is similar to the effects previously observed using other Immunosuppressive treatments (Sai P. et al., 1991 and Herold et al., 1996). For example, the results reported by Herold et al. (Herold et al., 1996) showed that anti-IFN-y mAb treatment only partially attenuated the MLDSTZ-induced elevation in blood glucose. A second factor that may explain the partial protective effect by A9-THC is that it is a relatively weak immunosuppressant. Moreover, the results obtained on Day 17 (data shown in Fig.lI-2 insert) further demonstrated that the attenuation of MLDSTZ-induced diabetes by 11-day treatment of Ag-THC is transient, which is similar to the effects produced by other immunosuppressive treatments. The termination of A9- 80 THC treatment on Day 11 may allow for the restoration of immune competence and subsequent infiltration of T-lymphocytes into the damaged islet. Further studies will be required to more fully elucidate the reason why a permanent protection was not achieved by Ag-THC treatment in this model. It is widely established that different mouse strains exhibit markedly different sensitivity to STZ treatment. For comparative purposes studies were also performed in BGC3F1 mice, a mouse strain not previously utilized in the MLDSTZ model of diabetes. In contrast to CD-1 mice, B6C3F1 mice exhibited minimum islet inflammation after MLDSTZ treatment (Fig.lI-4). In addition, the hyperglycemia was less pronounced after MLDSTZ treatment and the protection by Ag-THC was also less extensive than that observed in CD-1 mice. The decreased sensitivity of the B6C3F1 mice to MLDSTZ-induced hyperglycemia and loss of pancreatic insulin (Fig.lI-2, Fig.ll-3), as compared to CD-1 mice, can be explained in part by a less robust immune response in the pancreas (Fig.ll-4). However, the mild diabetic state produced in MLDSTZ-treated B6CBF1 mice which occurred in the absence of insulitis may reflect the direct cytotoxic action of MLDSTZ on pancreatic beta-cells. The premise that the direct toxic effects of STZ make a substantial contribution to the MLDSTZ model is further supported by the results of Gerling et al. from studies demonstrating that MLDSTZ was able to induce diabetes in NOD immune incompetent scid/scid mice (Gerling et al., 1994). Based on their results, lymphocytic infiltration was not observed and therefore was not required for diabetes development in those animals. The hyperglycemia induced in NOD scid/scid mice by MLDSTZ is likely due to direct 81 toxicity of STZ on beta-cells. Accordingly, these observations from different types of experiments suggest that sensitivity of beta-cells to the direct (i.e. non- immune) toxicity of STZ can elicit diabetogenic responses from laboratory animals given MLDSTZ treatment. In conclusion these studies strongly suggest that the mechanism responsible for MLDSTZ-Induced autoimmune diabetes involves at least two major elements. One element is associated with a robust immune response that is mounted against pancreatic cells. Most likely, it is the autoimmune component that is most effectively modulated by A9-THC treatment as suggested by our results in CD-1 mice. Of the two mouse strains examined, CD-1 mice exhibited the greatest magnitude of insulitis, the highest level of hyperglycemia and also the greatest magnitude of protection from Ag-THC co-administration. The second element appears to be more closely associated with the direct toxic action of STZ. This component of STZ-mediated diabetes is less sensitive to modulation by Ag-THC as suggested by our studies in BBC3F1 mice. These studies showed that 86C3F1 mice exhibited significant hyperglycemia and pancreatic insulin loss but very mild inflammation in pancreatic islets after MLDSTZ treatment and also were relatively insensitive to the protective actions of Ag-THC. The data presented in this report and by others, as discussed above, permit speculation regarding the degree of participation by the direct and the immune-based elements of MLDSTZ-induced diabetes in CD-1 mice. It appears likely that as much as one-half of the diabetogenic effects of MLDSTZ may be attributed to the direct, non-immune related toxic actions that are known to be characteristic of 82 STZ treatment in laboratory animals. For this reason use of the MLDSTZ model is appropriate for investigation of chemical-induced autoimmune response directed against the endocrine pancreas but it may be less appropriate for explanation of genetically based autoimmune diabetes. 83 CHAPTER III CANNABINOID RECEPTOR CB1-MEDIATED INSULIN RELEASE FROM ISOLATED PANCREATIC ISLETS AND RINM5F CELLS 84 lll.1.Summary Ag-Tetrahydrocannabinol (Ag-THC) modulates a wide variety of biological functions. Among these, it has been reported that Ag-THC treatment of rat pancreatic islets stimulated the release of insulin. The objective of the present studies was to further characterize the molecular mechanism responsible for A9- THC-stimulated insulin release and to determine whether this biological activity is mediated through cannabinoid receptors. The present studies demonstrate that Ag-THC treatment stimulated insulin release from isolated mouse pancreatic islets and from the rat-derived insulin producing RINm5F cells. Closely associated with Ag-THC-stimulated insulin release was a rapid but transient elevation in intracellular calcium. Interestingly, the calcium channel blocker, verapamil abrogated both the Ag-THC-induced elevation in intracellular calcium and release of insulin. In contrast, pretreatment with either thapsigargin or ryanodine, as inhibitors of calcium release from intracellular calcium stores, did not alter the elevation of intracellular calcium by Ag-THC. Moreover, the inhibitor of CaM kinase II, K93, but not the PKC inhibitor, bisindolylmalemide, diminished Ag-THC-stimulated insulin release. Analysis of both primary pancreatic islets as well as RINm5F cells by RT-PCR identified mRNA expression for the central cannabinoid receptor, CB1. Interestingly, the CB1 receptor antagonist, SR141716A, blocked Ag-THC-stimulated insulin release from both isolated mouse islets and from RINm5F cells. In addition, pretreatment of RINm5F cells with SR141716A blocked the rise in Ag-THC-induced elevation in intracellular 85 calcium. Collectively, these results demonstrate that Ag-THC-stimulated insulin release is mediated through an elevation of intracellular calcium through the opening of calcium channels via a CB1-dependent mechanism. lll.2.lntroduction Stimulation of insulin secretion by pancreatic beta-cells in response to most secretagogues involves changes in intracellular calcium concentration [Ca2+]i ( Seri et al., 2000; Fischer et al.,1999; Maechler et al., 1999; Lange and Brandt, 1993). Glucose and other nutrients that stimulate insulin release produce an elevation in [Ca"”’]i via mechanisms that induce membrane depolarization and a calcium influx via the opening of voltage sensitive calcium channels and through the release of calcium from intracellular stores (Takasawa et al., 1998; Zawalich et al., 1998; Safayhi et al.,1997; Porksen et al., 1996; Tang et al., 1995). A variety of calcium-sensitive intracellular regulatory elements involved in the insulin secretory process have been identified. One of the most extensively characterized is the calcium/calmodulin dependent kinase, CaM kinase II. Numerous studies have demonstrated an increase in CaM kinase II activity under conditions where insulin release was induced (Mohlig et al., 1997; Norling et al., 1994). Likewise, the inhibition of CaM kinase II activity has been closely correlated to an inhibition of insulin release (Wenham et al., 1994). Collectively, CaM kinase II is widely established as an important transducer of calcium signaling and intimately involved in the phosphorylation of proteins that regulate insulin granual excytosis. 86 Ag-Tetrahydrocannabinol (Ag-THC), the primary psychoactive constituent of marihuana, modulates a wide variety of biological functions (Wenger et al., 1999; Voth and Schwartz, 1997; Smith et al., 1978) . Among these, it has been reported that Ag-THC treatment of isolated rat pancreatic islets stimulated the release of insulin (Laychock et al., 1986). Although the mechanism responsible for Ag-THC-stimulated insulin release in the aforementioned study was not established, it is notable that these studies were conducted prior to the identification and cloning of the two major forms of cannabinoid receptors, CB1 and C82 (Matsuda et al., 1990; Munro et al., 1993) . CB1 is predominantly expressed within the central nervous system (Howlett, 1998), but has also been identified in peripheral tissues including the lung, testis, hearts, spleen, and small intestine (Felder and Glass, 1998; Rice, 1997; Pertwee et al., 1996). The second and more recently characterized cannabinoid receptor subtype, C82, is not expressed In the brain but has been identified in a number of peripheral tissues with the immune system being best characterized (Schartz et al., 1997; Munro et al., 1993; Kaminski et al., 1992). It has now been established that certain cannabinoids mediate at least part of their biological activity through one or both of these cannabinoid receptors. Both CB1 and C82 are G-protein coupled receptors and have been demonstrated to modulate a number of different signal transducing elements including adenylate cyclase, ERK MAP kinases and most important to the present investigation intracellular Caz+ (Bouaboula et al., 1996; Condie et al., 1996; Schartz et al., 1992; Kaminski et al., 1994). The objectives of the present study were two-fold: 1) to investigate whether calcium-dependent 87 signaling pathways are responsible for Ag-THC-stimulated insulin release; and 2) to determine whether this biological activity is mediated through cannabinoid receptors. |II.3.Materials and methods lll.3.A.Materials All reagents were purchased from Sigma Chemical Co. (St. Louis, MO) unless otherwise identified. RPMI-1640 and other cell culture reagents were purchased from BRL Life Technologies (Gaitherburg, MD). Ag-THC and cannabidiol (CBD) were provided by the National Institute on Drug Abuse (National Institute of Health, Rockville, MD). Stock solutions of A9-THC and CBD were prepared in ethanol and added directly to the culture medium to achieve the desired experimental concentrations. Cannabinoid receptor CB1 antagonist SR1141716A and C82 antagonist SR144528 were also obtained from the National Institute on Dmg Abuse (National Institute of Health, Rockville, MD) and prepared in DMSO. RT-PCR reagents were purchased from Promega Co. (Madison, WI). Research grade collagenase from Clostn'dium histolyticum was obtained from Boehringer Mannheim (GmbH, Gemany). Fura-2 acetoxymethyl- ester (Fura-2/AM) was obtained from Molecular Probes (Eugene, OR). The CaM kinase II inhibitor (KN93) was purchased from Calbiochem-Novabiochem Corp. (La Jolla, CA). lll.3.B.Preparation of mouse pancreatic islets 88 Mouse pancreatic islets were prepared from 8 to 9 week old male CD-1 mice (Charles River Laboratory, Dortage, Ml). Upon arrival, mice were randomly transferred to standard filter topped cages containing sawdust bedding. Five mice per cage were housed in a pathogen-free facility, which was maintained at 21-24°C constant temperature, 40~60% relative humidity and a 12 hour light/dark cycle. Mice were provided standard laboratory animal chow (Purina Certified Laboratory Chow) and water ad Iibitum. Isolated pancreatic islets from CD-1 mice were prepared by collagenase digestion (Gotoh et al., 1985) and were maintained in culture overnight in complete RPMl-1640 medium supplemented with 10% heat-inactivated fetal calf serum, 200 ug/ml penicillin and 200 mU/ml streptomycin to recover from the isolation procedure. lll.3.C.Cells Rat insulinoma-derived RINm5F cells were generously provided by Dr. Paolo Meda (Geneva, Switherland). The cells were cultured in a humidified atmosphere of 95% O2 and 5% CO2 at 37°C in complete RPMl-1640 medium which is supplemented with 10% heat-inactivated fetal calf serum, 200 pg/ml penicillin and 200 mU/ml streptomycin (GIBCO, Grand Island, NY). The medium was changed every other day, and the cells (passage 40-60) were utilized for experiments on Day 5 after plating. |II.3.D. Measurement of insulin release Cultured pancreatic islets were distributed into polypropylene centrifuge 89 16‘ alic vel can we the de IIC Fl: I6 aI tubes in batches of 10 islets per treatment group. The islets were preincubated for 15 min with 1ml of oxygenated Krebs-Ringer bicarbonate (KREB) buffer supplemented with 11.4 mM glucose and 0.01% bovine serum albumin (BSA) (fraction V; fatty acid free) at 37°C in a COzincubator (95% O2 and 5%). Similarly, RINm5F cells were transferred into 6-well culture plates containing fresh RPMI- 1640. The release of insulin from islets was determined by measuring insulin in aliquots of culture supernatant sampled after a 30 min incubation with A9-THC or vehicle (0.1% ethanol) unless indicated otherwise. In experiments where cannabinoid receptor antagonists, calcium channel blockers or kinase inhibitors were employed, the cultures were preincubated with these respective agents, or the vehicle control (0.1 % DMSO), for 30 min prior to treatment with Ag-THC. Media insulin was assayed using a radioimmunoassay as previously described (Fischer et al., 1996). An aliquot of KREB buffer or culture medium was removed and stored at —20°C until assayed. Release of insulin was normalized to milligrams of total cellular DNA content (measurement described in Fischer et al., 1996). Viability of cells was assessed by trypan blue exclusion. In addition, the release of RINm5F cells of lactate dehydrogenase (LDH) into the medium was also used as the further indicator of cytotoxicity (measurement according to the method of Bergmeyer and Bemt, 1974). |II.3.E. Determination of cannabinoid receptor mRNA expression Trizol reagent (3ml) (Life Technologies, Grand Island, NY) was added to 90 500 to1000 cultured pancreatic islets or 1x107 RINm5F cells for total RNA isolation. All isolated RNA samples were DNAse treated to eliminate any potential genomic DNA contamination prior to the RT-PCR. Briefly, RNA were reverse-transcribed simultaneously into cDNA in 20 pl reaction buffer containing oligo (dT)15 and Molony Murine Leukemia Virus reverse transcriptase. Then a PCR mix consisting of 10x PCR buffer (20 mM ammonium sulfate, 67 mMTris, 6.7 pMEDTA, 8 mg/ml BSA, and 5 mM 2-mercaptoethanol), 4 mM MgCIz, 6 pmol each of forward and reverse primers, and 1.25 units of Taq DNA polymerase was added to the cDNA samples. Samples were then heated to 94°C for 4 min and cycled at a 94°C denaturing step for 15 sec, a 59°C annealing step for 30 sec, and a 72°C elongation step for 30 sec, after which an additional extension step at 72°C for 5 min was included. Specifically, for the detection of CB1 and C82 transcripts, 400 ng RNA and 35 amplification cycle. Primer sequences for mouse CB1 are: fonlvard primer GGATATCTGGAGGAACTGGC, reverse primer, GAGCTCATTGAATGCTTGGC; for mouse CB2 primer: froward primer, CAGTACACCTGCCACAAAGGA, reverse primer, GTGTGACCTTCTCTGCAGA CA. Primer sequences for rat CB1 are: fonNard primer CTGTGGAGGAACGGAT ATGC, reverse primer, TTGAAGTCATGCTTGAGCGC; for mouse C82 primer: froward primer, GAACCTGCCAGTACCAACAAG, reverse primer, CTTCGACTG TGTGCAACTCGA. The RT-PCR product was determined by densitometric analysis of ethdium bromide-stained agarose gel (3%, Nusieve; agarose=3:1) using the Gel Doc 1000 analysis system (BioRad Laboratories Inc., Herculese, CA). 91 lll.3.F.Measurement of [Caz‘L [Ca2+], was measured in Fura-2/AM loaded cells using a SPEX FIuorolog-2 Spectrofluorometer (SPEX Industries, Inc., Edison, NJ.) RINm5F cells were aliquoted at density of 2x104 cells/ml in 25 cm2 flasks. After 5 days of culture, the cells were detached from the flask by treatment with trypsin, washed twice with a modified KREB buffer, adjusted to a density of 1 X 106 cells/ml and incubated with 1uM Fura-2/AM at 37°C. After 30min incubation, the loading buffer was removed and the suspended cells were rinsed twice with fresh KREB buffer. Fura-2/AM-loaded cells (1 X 10" cells/ml) were added to a cuvette and the temporal changes in [Ca2+],-induced fluorescence by various chemical treatment were measured. Fluorescence emission at 505 nm was monitored at room temperature with constant stirring, using a dual wavelength spectrofluorometer system with excitation at 340 and 380 nm. At the end of scanning, 0.1% Triton X-100 was added to determine the maximum fluorescence (Rmax) and then 0.015M EGTA was added to obtain minimum fluorescence (Rmin). These measurements were used in computer-based calculations of [Ca2+], to obtain the representative graphs. The [Ca”], was calculated from fluorescence intensity readings using the following equation: [Ca2*]i = Kd*Q(R-Rm,,,)/(Rma,,-R). R is the ratio of emission intensities at 340 and 380 nm excitation. Kd is the dissociation constant of the Ca2*/Fura-2 complex and equal to 1.45 X107; and Q is the ratio of the 380 nm fluorescence under conditions of minimum and maximum [Ca2*] , conditions (Shao et al. 1998). 92 lll.3.G.Statistical analysis Data are presented as the mean :I: SEM from at lease three experiments conducted in triplicate (n=9). Treatment groups were compared with the vehicle control groups using an analysis of variance and Dunnett's two-tailed t-test, with p< 0.05 considered statistically significant. III.4.Results Ill.4.A.Stimulation of insulin release from mouse pancreatic islets and RINm5F cells by A°-THC treatment Mouse pancreatic islets and insulin-producing RINm5F cells were employed to examine the direct effects of Ag-THC treatment on insulin release. Figure Ill-1 shows that a 30 min treatment with Ag-THC stimulated the release of insulin from both mouse islets (Fig.lll-1A) and RINm5F cells (Fig.llI-1 B) in a concentration-dependent manner during a 30-min treatment period. At the highest concentration tested, Ag-THC (15 pM) produced an approximately 3-fold increase in insulin release by RINm5F cells, as compared to control. The treatment of RINm5F cells or pancreatic islets with Ag-THC at concentrations equal or lower than 15pM was not cytotoxic as assessed by release of LDH into culture mediums and/or trypan blue exclusion. Ill.4.B.A°-THC-induced increase in [Ca”]. in RINm5F cells One of the proximal events governing insulin release is a rapid increase in 93 400 - e I. 2 .2 c 300 ‘ a U) 5 g 200 I N 2 O a: T I: 100 I '5 ID 5 0 I T NA VH 0.2 5 15 Ag-THC (uM) Treatment Group Figure Ill-1A. Effect of Ag-THC on insulin release from mouse pancreatic islets. Islets were incubated in KREB buffer containing: no treatment (NA); vehicle (VH); and Ag-THC as indicated. The insulin release into the medium was assayed over a 30 min period and normalized per 50 islets. The results are expressed as the mean t SE, n=9. A significant difference of p<0.05 is indicated where “*" denotes mean values that are different compared to the VH control group. 94 15- 12- Insulin Release (nglpg DNA) \\\\\\\\\§t z < PAL 0 M o N o N A'-THC (uM) Treatment Figure Ill-1 B. Effect of A9-THC on insulin release from RINm5F cells. Cultures were incubated with: no treatment (NA); vehicle (VH); and A9-THC as Indicated. The insulin release into the medium was assayed over a 30 min period and normalized to DNA content. The results are expressed as the mean i SE, n=9. A significant difference of p<0.05 is indicated where “*" denotes mean values that are different compared to the VH group. 95 [Ca2+],. To determine whether AQ-THC-stimulated insulin release was being mediated through a rise in [Ca2+],, [Ca2+]i was measured in Fura-2/AM-Ioaded RINm5F cells. In control cells (Fig.lII-2), the free calcium level did not rise upon addition of vehicle (0.1% ethanol) and remained constant throughout the experimental period. In Ag-THC-treated cells (Fig.llI-2), calculated [Ca""‘ji was elevated in approximately 50 sec and exhibited a maximal 3-fold increase within 300 sec after drug addition. The elevation in [Ca2*]i was rapid and sustained, persisting for more than 300 sec. No further increase in [Ca2*]i was observed within a 30 min time period (data not shown). lll.4.C.Attenuation of A’-THC-induced Increase in [Caz‘]I and insulin release by the calcium channel blocker verapamil To further determine whether insulin release stimulated by Ag-THC was associated with a rise in [Ca2*]i via influx through plasma membrane associated calcium channels, the calcium channel blocker verapamil was utilized. The results in Figure Ill-3A show that verapamil (40 pM) pretreatment abrogated the increase of [Cam]i induced by A9-THC in RINm5F cells. In a separate experiment, verapamil completely blocked the insulin release stimulated by A9-THC in RINm5F cells (Fig.lll-3B). Verapamil treatment alone neither altered basal insulin release (Fig.lll-3B) nor changed the basal level of [Ca2+], (Fig.lII-3A). These results suggest that the source for the increase of [Ca2+]i induced by Ag-THC is extracellular. 96 200 I TIL .3 an. 1 50 3 "- flux!” M... 9 100 « wwwwy A JHC 50 WMWM‘JI‘.l‘MmLWWMMWMf-fihWWMA VH Calculated [Ca2+]i (10'8M) o 160 260 330 450 560 600 Time (sec) Figure Ill-2. Effect of Ag-THC on [Ca2*]i in RINm5F cells. AQ-THC or vehicle (VH) were administered at the arrow to the Fura-2/AM loaded cell suspension. [Ca"’*]i was measured and determined as described in Materials and Methods. Data shown are the representative of five experiments. 97 IIMwI 5° I I Verapamil Ag-THC 250‘ $2 200, O 1- 2; f 150‘ VH A9-TI-Ic N 2. I I ‘U 100 s 2 3 0 a U f I fi 0 100 200 300 400 500 600 Time (sec) Figure III-3A. Effect of calcium channel blocker verapamil on AQ-THC-induced increase in [Caz*]i in RINm5F cells. Fura-2/AM loaded cells were treated with 15 pM of A9-THC in the presence of vehicle (VH) or 40 pM verapamil. Arrows indicate additions of treatments. [Ca2*]i was measured and determined as described in Materials and Methods. Data shown are the representative of three experiments. 98 2 a z 25 w . D E: OpM Verapamil l C» - 40 pM Verapamil 2 20 . g a V 15 . . I d) b 32 1o - ‘7 b b c = 5 < 3 In C _ 0 ‘ _ _ _ VH 0.2 5 15 Ag-THCQIM) Treatment Group Figure III-38. Effect of calcium Channel blocker verapamil on the stimulation of insulin release by A9-THC in RINm5F cells. Cultures were incubated with verapamil for 30 min prior to exposure to vehicle (VH) or A9-THC treatment for additional 30 min. The insulin release into the medium was assayed and normalized to DNA content. Data are expressed as the mean t SE, n=9. A significant difference of p<0.05 is indicated where “a” denotes mean values that are different compared to the corresponding vehicle control receiving no verapamil treatment; and “b” denotes mean values that are different compared to the corresponding control receiving verapamil treatment. 99 lll.4.D.Thapsigargin and ryanodine exert no effect on the elevation in [Cap]l induced by A°-THC To confirm that the rise in [Ca2+], induced by Ag-THC was not due to the mobilization of calcium from intracellular calcium stores, additional studies were performed utilizing thapsigargin. Control experiments confirmed the ability of thapsigargin to deplete intracellular calcium stores and inhibit the ATP- dependent Ca2+ reuptake as evidenced by the inability of carbachol, a known releaser of Ca2*-stored in the ER, to increase [Ca2+]. after thapsigargin treatment. Similarly, thapsigargin treatment alone rapidly increased [Ca2*].. Interestingly, thapsigargin did not alter the increase of [Ca2+]i induced by Ag-THC in RINm5F cells (Fig.lIl-4A). In a separate experiment, the increase in [Ca2”]i induced by A9- THC was not altered in the presence of ryanodine (10 pM) pretreatment (Fig.lll- 4B). The latter is able to block Ca2+ release from intracellular Ca2+ stores through ryanodine-sensitive Channels. These studies confirm that source for the Increase of [Ca2+], induced by Ag-THC is extracellular. lll.4.E.Effects of CaM kinase II inhibitor KN93 and PKC inhibitor . bisindolylmalemide on the stimulation of insulin release by A9-THC Two calcium regulated protein kinases, CaM kinase II and PKC, are well established as being critically involved in the regulation of insulin release (Gromada et al., 1999; Miura et al., 1998; Gregersen et al., 2000b; Tabuchi et al., 2000). To further examine the putative involvement of CaM kinase II and/or 100 A. c_ VH Carbachol I 1x1o‘M Carbachol Calculated [Cam]I F———)I m 0 100 200 300 400 500 600 0 100 200 300 400 500 600 Time (sec) Time (sec) Figure Ill-4A. Effect of thapsigargin on Ag-THC-induced increase in [Ca2“]i in RINm5F cells. The ability of 2 pM thapsigargin (TG) to deplete intracellular calcium stores and inhibit the ATP-dependent Ca2+ reuptake was confirmed by treatment with 200 nM carbachol (A: vehicle, B: thapsigargin). In separate experiments, fura-2/AM loaded cells were treated with 15 pM AQ-THC in the presence of C: vehicle (VH) or D: 2 pM thapsigargin (TG). Arrows indicate additions of treatments. [Ca2+], was measured and determined as described in Materials and Methods. Data shown are the representative of three experiments. 101 200 . E . °.° ' " I t o 150‘ I “I; :2:- I + ,' mm . 9.. 1004 I “g V“ A9-THC .l E ‘ I A' a 3 5 INIWNIIMWJ (U 0 I Ryanodine I A9-THC d 0 1 00 200 300 400 500 600 Time (sec) Figure III-4B. Effect of ryanodine on Ag-THC-induced increase in [Ca2+], in RINm5F cells. Fura-2/AM loaded cells were treated with 15 pM Ag-THC in the presence of vehicle (VH) or 10 pM ryanodine. Arrows indicate additions of treatments. [Ca2*]i was measured and determined as described in Materials and Methods. Data shown are the representative of three experiments. 102 PKC in A9-THC-stimulated insulin release, experiments were performed utilizing selective inhibitors of these two enzymes. As illustrated in Fig-III.5A, KN93, an inhibitor for CaM kinase II, completely inhibited the stimulation of insulin release by A9-THC whereas KN93 by itself did not Change the basal level (no Ag-THC treatment) of insulin release. In contrast, the PKC inhibitor bisindolylmaleimide produced no effect on Ag-THC-stimulated insulin release (Fig-lll.5B). As a control to assess the efficiency and extent of PKC down-regulation by bisindolylmaleimide, TPA-induced stimulation of insulin release (Tang et al., 1998) was measured. TPA-induced stimulation of insulin release in the presence of bisindolylmaleimide was reduced by more than 50%. III.4.F.Identification of cannabinoid receptor mRNA expression in isolated pancreatic islets and RINm5F cells Although cannabinoid receptor expression has been identified in a variety of cell types and tissues, insulin-producing cells have not been previously examined for either CB1 or CB2. To investigate the potential involvement of cannabinoid receptors in the insulinotropic action of Ag-THC, the expression of both CB1 and CB2 mRNA was examined in CD-1 mouse pancreatic islets and RINm5F cells using RT—PCR analysis (Fig-Ill.6). Figure III-6 involvement of cannabinoid receptors in the insulinotropic action of A9-THC, the expression of both CB1 and CB2 mRNA was examined in 00-1 mouse pancreatic islets and RINm5F cells using RT-PCR analysis (Fig-Ill.6). Figure Ill-6 shows that CD-1 mouse pancreatic islets express mRNA for the CB1 receptor but not the CB2 103 g 25 . 1:1 0 “M Ag-THC o 15pM A9'THC a 5:1 20 . \ c» 5 a: 15 ‘ g b % 10 - a. % c / E o , / VH 10 5 1 KN93 (pM) Treatment Figure Ill-5A. Inhibition of AQ-THC-stimulated insulin release from RINm5F cells by the CaM kinase II inhibitor, KN93. RINm5F cellswere incubated with KN93 or vehicle (VH) for 30 min prior to exposure to Ag-THC treatment for additional 30 min. The insulin release into the medium was assayed and normalized to DNA content. The results are expressed as the mean i SE, n=9. A significant difference of p<0.05 is indicated where “a" denotes mean values that are different compared to the vehicle control receiving no KN93 treatment; and “b” denotes mean values that are different compared to the corresponding control receiving Ag-THC treatment. 104 25 ‘ :1 VH 15pM A°-TI-IC ’ /’ 50nM TPA 20' 15. y/ / S 10‘ \\ // ///,. 0.1% DMSO 2uM BIS Treatment Group 9% O Insulin Release (nglug DNA) Figure Ill-5B. Effect of bisindolylmaleimide (BIS), a PKC inhibitor, on Ag-THC- stimulated insulin release from RINm5F cells. RINm5F cells were incubated with bisindolylmaleimide or 0.1% DMSO for 30 min prior to exposure to vehicle (VH), TPA, or Ag-THC for additional 30 min. The insulin release into the medium was assayed and normalized to DNA content. Data are expressed as the mean i SE, n=9. A significant difference of p<0.05 is indicated where “a” denotes mean values that are different compared to the corresponding vehicle receiving no bisindolylmaleimide treatment; “b” denotes mean values that are different compared to the corresponding vehicle receiving bisindolylmaleimide treatment; and “c” denotes mean values that are different compared to the corresponding control (0.1% DMSO) receiving TPA treatment. 105 receptor. In contrast, mRNA for both CB1 and CBZ was readily detected in RINm5F cells. Ill.4.G.Effects of cannabinoid receptor antagonists on A°-THC- stimulated insulin release In light of the Identification of CB1 and C82 mRNA expression by insulin producing cells, additional studies were performed to investigate the involvement of cannabinoid receptors in the insulinotropic actions of Ag-THC. For these experiments, the CB1 selective antagonist, SR141716A, and the C82 selective antagonist, SR144528, were employed. Pretreatment of the mouse pancreatic islets (Fig.llI-7A) and RINm5F cells (Fig.lll-7B) with the CB1 antagonist, SR141716A (10 pM) inhibited AQ-THC-stimulated insulin release. In a separate experiment, SR144528 (10 pM), a CB2 receptor-specific antagonist, did not affect Ag-THC (15 pM)-stimulated insulin release from RINm5F cells (Fig.lll-7C). As shown in figures III-7, neither 10 uM of SR 141716A nor 10 pM of SR144528 alone altered basal insulin release (no Ag-THC treatment). Furthermore, cannabidiol, a plant-derived cannabinoid with low affinity for both CB1 and CB2, was unable to induce insulin release from RINm5F cells at the same concentration that Ag-THC exhibited a maximum release of insulin (Fig.llI-7D). lll.4.H.Effects of cannabinoid receptor antagonists on A°-THC- induced [Ca2*]. increase in RINm5F cells 106 Islet Islet Spleen RINm5Fcells CBZ—_* CB1——* GAPDH Lane 1 2 3 4 5 Figure Ill-6. RT-PCR analysis of CB1 receptor mRNA and C82 receptor mRNA expression in the mouse pancreatic islets and RINm5F cells. RT-PCR analysis was performed as described under “Materials and Methods". Lane1: mouse CB1; ‘ lane 2: mouse CB2; lane 3: mouse CB1; lane 4: rat CB1; lane 5: rat CB2. Results are representative of three separate experiments. 107 400 A 10 fl 2 m ._ . a o 300 a In ‘ """" é \ , a: p, 5 j a, 200- U) (B 2 a: n: 100- IE 3 m E 0 VI-I THC 0.001 0.01 0.1 1 10 10 SR (pM)+A9-THC SR (0M) Treatment Figure Ill-7A. Effect of SR141716A on Ag-THC-stimulated insulin release from mouse pancreatic islets. Islets were pretreated with SR141716A (SR) for 30 min prior to exposure to 15 pM Ag-THC as indicated. Additional groups were employed as control (VH: 0.1% DMSO and 0.1% ethanol; THC: 0.1%DMSO and 15 pM Ag-THC; SR10: 10pM SR141716A and 0.1% ethanol). The insulin release into the medium was assayed and normalized per 50 Islets. Data are expressed as the mean t SE, n=9. A significant difference of p<0.05 is indicated where “a” denotes mean values that are different compared to the VH group; and “b" denotes mean values that are different compared to the THC group. 108 2 15' CI OpMSR z -0.1p.MSR D 12, 1pMSR CI 3 \ o: 5 9. 0: a m a g l E b .s g E In E .2 5 A9-THC(uM) Treatment group Figure Ill-7B. Effect of SR141716A on Ag-THC-stimulated insulin release from RINm5F cells. Cells were preincubated with CB1 antagonist SR141716A (SR) for 30 min prior to exposure to Ag-THC or vehicle (VH) treatment as indicated. The insulin release into the medium was assayed and normalized to DNA content. Data are expressed as the mean i SE, n=9. A significant difference of p<0.05 is indicated where “a” denotes mean values that are different compared to the corresponding VH group receiving no Ag-THC; and “b” denotes mean values that are different compared to the corresponding 0 pM SR group receiving same concentrations of AQ-THC. 109 15 ‘ 1:: VH :f — 150M A’JHC 2 D 12 ' * o: E E 9‘ o 8 .42 6‘ a: 0: .E 3 : ‘5 in c — 0 0 10 SR 144528 (uM) Figure Ill-7C. Effect of SR144528 on AQ-THC-stimulated insulin release from RINm5F cells. Cells were preincubated with C82 antagonist SR144528 for 30 min prior to exposure to Ag-THC or vehicle (VH) treatment. The insulin release into the medium was assayed and normalized to DNA content. Data are expressed as the mean 1- SE, n=9. A significant difference of p<0.05 is indicated “*I! where denotes mean values that are different compared to the corresponding VH group. 110 .t 0| < z D12- :31 3 \ ow * 5 9- o In 10 .9.’ 6- o 0: C 1.: 3. 3 .E 0 e. . 9 NA VH CBD A-THC Treatment Group Figure III-7D. Effect of cannabidiol on insulin release from RINm5F cells. Cultures were incubated with: no treatment (NA); vehicle (VH); 15 pM cannabidiol (CBD) and 15 pM Ag-THC as indicated. The insulin release into the medium was assayed over a 30 min period and normalized to DNA content. Data are expressed as the mean : SE, n=9. A significant difference of p<0.05 is 11*” indicated where denotes mean values that are different compared to the corresponding VH group. 111 ”OI E 00 '9 2001 S +.:= N 150. N 9" V" A9 TH '3 100. I (I a l! 3 2 50 fl 0 SR A9-THC 0 100 200 300 400 500 600 Time (sec) Figure Ill-8. Effect of SR141716A on AQ-THC-induced increase in [Ca”], in RINm5F cells. Fura-2/AM loaded cells were treated with 15 pM of A9-THC in the presence of vehicle (VH) or 1 pM SR141716A (SR). Arrows indicate additions of treatments. [Ca2+], was measured and determined as described in Materials and Methods. Data shown are the representative of three experiments. 112 The role of the CB1 receptor in the elevation of [Ca2+], induced by Ag-THC was examined using CB1 receptor antagonist SR141716A. At concentrations of 1 uM SR141716A blocked the rise in [Ca2+], induced by Ag-THC (15 pM) (Fig.lll- 8). SR141716A (1 and 10 pM) alone did not alter basal levels of [Ca2*]i (data not shown). lll.5.Discussion Results from the present study demonstrate that Ag-THC treatment produced a cannabinoid receptor-mediated increase in [Ca”], and a marked increase in insulin release from isolated mouse pancreatic islets and rat insulinoma-derived RINm5F cells (summarized in Fig.lII-9). These results are consistent with and extend those in a previous report which indicated that insulin was released from rat pancreatic islets treated with the cannabinoid (Laychock et al., 1986). Based on their report, Ag-THC was able to increase both high glucose (8.5 mM) stimulated and basal (glucose, 2.8 mM) insulin release from isolated islets. RINm5F cells are known to be unresponsive to high concentrations of glucose as a stimulant for insulin release (Halban et al., 1983), but do release insulin as a result of treatment with other insulin secretagogues, eg. amino acids, sulfonylurea drugs (Laychock, 1998; Leiers et al., 2000). The stimulation of insulin release from in vitro preparations by Ag-THC treatment appears to be a direct action on insulin-producing cells, which is independent of high glucose concentrations. A comparison of the concentration-response curves for AQ-THC- 113 induced insulin release obtained in mouse islets and RINm5F cells in the present study with that obtained using rat pancreatic islets (Laychock et al.,1986) indicates that they are similar. The lowest concentration of Ag-THC eliciting insulin release is in the 200-500 nM range with increasing hormone released as the cannabinoid concentration was increased to 15-20 pM. This similarity of response suggests a common mechanism of action for A9-THC in each cellular preparation. A previous report (Laychock et al., 1986) showed an involvement of arachidonic acid (AA) release and possibly Iipoxygenase products may be involved in Ag-THC-stimulated insulin release. In the present study, the involvement of several other, important intracellular signaling pathways for insulin release were investigated to provide additional insight into the mechanism of action by Ag-THC. The results demonstrated a marked elevation of [Ca2*]i is Closely associated with Ag-THC-stimulated insulin release in RINm5F cells. This finding is consistent with the accepted view that [Ca2+], is an important intracellular messenger regulating insulin release from pancreatic beta-cells ( Seri et al., 2000; Fischer et al.,1999; Maechler et al., 1999; Lange and Brandt, 1993). The primary source of the increase in [Ca2+], appears to be the opening of verapamil-sensitive calcium channels presumably on the plasma membrane of the cell. Inhibiting the rise in [Ca2*]i with verapamil pretreatment (Fig.lll-3A) is consistent with the demonstrated inhibitory action of the calcium channel blocker on Ag-THC-induced insulin release from RINm5F cells (Fig.lll-3B) and from 114 isolated mouse pancreatic islets (data not shown). This result is also concordant with data indicating an influx of extracellular calcium through voltage-dependent L-type channels or voltage-independent Channels is required for insulin release stimulated by many nutrient and non-nutrient secretagogues (Safaihi et al., 1997; Boyd,1992) Ag-THC-induced increases in [Ca2*]i could also occur by mobilization of the ion from major intracellular stores such as those in the endoplasmic reticulum (ER) of insulin producing cells. Pretreatment of RINm5F cells with two different inhibitors of Ca” mobilization from the ER, thapsigargin and high ryanodine concentrations, did not block the Ag-THC-induced rise in [Ca2+],. This result is consistent with little or no role for the ER, the major intracellular storage site for Ca”, as a source of the AQ-THC-induced rise in [Ca”],. Other possible sources of the cannabinoid-induced rise in [Ca2+],, including mitochondrial and nuclear stores, will be investigated in future experiments directed toward further elucidation of the calcium signaling pathway. Previously published reports indicate that AQ-THC treatment can alter [Ca2+], in a variety of different cell types. Ovenlvhelmingly, the majority of these studies have demonstrated an inhibitory effect of Ag-THC on the opening of several types of calcium channels (Twitchell et al., 1997; Pan et al., 1996; Shen and Thayer, 1998). In addition, Ag-THC caused a concentration dependent inhibition of calcium mobilization elicited by caerulein in the exocrine pancreas (Laychock et al., 1988). Several reports have also shown that a rise in [Ca”], is 115 observed upon A9-THC treatment In several cell types or cell line. For example, a treatment-related increase of [Ca2+]i in DDT1MF-2 smooth muscle cells has been observed (Filipeanu et al., 1997) and neuroblastoma x glioma hybrid cell NG108- 15 cells and N18TG2 cells showed a rapid increase in [Ca”], (Sugiura et al., 1997). Considering these and other reports, it is likely that cell and/or species specific effects may be prominent In the calcium signaling pathways responsive to Ag-THC. Increases In [Ca2*]i after Ag-THC exposure can be expected to activate a variety of calcium-dependent enzymes. Among those, CaM kinase II (Easom, 1999) and PKC (Howell et al., 1994) have been extensively studied and reported to be activated in RINm5F cells In response to known insulin secretagogues (Gromada et al., 1999; Miura et al., 1998; Gregersen et al., 2000b; Tabuchi et al., 2000). By using selective inhibitors of these two kinases, KN93 for CaM kinase II and bisindolylmaleimide for PKC, the results of the present study suggest a possible role of CaM kinase II in the calcium-dependent signal transduction mechanism involved in Ag-THC-stimulated insulin release and rule out the possible involvement of calcium-dependent PKC (Fig.lll-SB). While there is evidence showing that the degree of specificity of KN-93 can be questioned, results from preliminary experiments indicate that KN-93 when used at 10 pM did not block a Ag-THC-induced increase in [Ca"’*]i (data not shown) but as shown in Fig.llI-5A did inhibit the cannabinoid-induced insulin release (Fig.lII-5A). Thus, attributing the inhibitory effect of KN-93 on insulin release to a calcium channel 116 blocking action, similar to that possessed by another CaM kinase inhibitor KN- 62, is not possible (Li et al.,1992). To date, the tissue and cell type distribution of CB1 and CB2 has not yet been comprehensively characterized. The results of the present study have clearly demonstrated mRNA expression of CB1 cannabinoid receptor in isolated mouse pancreatic islets and RINm5F cells by RT—PCR analysis (Fig.lll-6). These findings are the first to identify pancreatic islet-cells as another periphery location for CB1 receptor/ligand interactions. Notably, mRNA expression for the CB2 receptor was detected in RINm5F cells but not in mouse pancreatic islets by RT- PCR analysis (Fig.lll-6). The reason why RINm5F cells but not islet cells possess CB2 receptor mRNA may be a reflection of either the transformed phenotype of the Cloned cells or differences among rodent species. In addition, the possibility of CB1 or CB2 expression In endocrine cells within islets other than the beta- cells cannot be determined from present studies and will require further examination. Based on the identification of CB1 transcripts in mouse pancreatic islets and CB1 and CBZ transcripts in RINm5F cells, further studies were performed to examine the potential involvement of cannabinoid receptors In Ag-THC- stimulated Insulin release. One striking observation was that the CB1 antagonist, SR141716A, antagonized insulin release Induced by Ag-THC in both mouse islets and in RINm5F cells. Moreover the antagonism by SR141716A of the A9- THC-induced insulin response in both cell preparations was concentration dependent, suggesting an involvement of CB1 receptor. Additional experiments 117 also demonstrated that SR141716A was capable of completely abolishing the A9- THC—induced increase In [Ca"’*]i in RINm5F cells, further supporting our premise that the induction of insulin release by pancreatic beta-cells in the presence of cannabinoid treatment is critically linked to the elevation of [Ca2+],. Although it is not possible to discern from the present studies whether CB1 directly or indirectly couples to calcium channels in the pancreas-derived cell preparations, nevertheless the results suggest that the influx of calcium that is induced by A9- THC is regulated through a CB1-dependent mechanism. To our knowledge, this is the first report demonstrating that a cannabinoid ligand Induces calcium influx through a CB1-dependent mechanism. Experiments employing the C32 antagonist, SR144528, showed no effect on Ag-THC-stimulated insulin release from RINm5F cells (Fig.lll-7C) ruling out the involvement of CB2 receptors. The findings from this present Investigation allow for some interesting but speculative hypothesis to explain previously reported observations. From a recent Investigation conducted in this laboratory it was reported that CD-1 mice treated daily with AQ-THC exhibited protection from streptozotocin-Induced diabetes (Li et al.,1999). This protective effect was partially due to a suppression of the Immune-mediated destruction of beta-cells initiated by multiple low dose administration of streptozotocin. It Is tempting to speculate that the secretagogue-like effect of A9-THC reported here on insulin-producing cells in vitro could also partly account for the reduction in streptozotocin-induced hyperglycemia observed in this model in association with Ag-THC administration. 118 .353 .5de on. E0: $.39 05 .3 9.8883 mm. 9955mm. 05.95% Eon:oaou-E:_o_mo m 59.95 030.9 5.3:. um~m_:_E=m-OI..-m< BEBE .9309 50 6.... 2:9”. ...,umme.x.2mo .. All =2 35:. I-11-----, m ,=__=me_, .. 119 CHAPTER IV OVERALL SUMMARY AND CONCLUSION 120 Diabetes mellitus is recognized as the world's most common metabolic disorder, affecting people worldwide and of all age groups. The severity of diabetes is reflected both in its prevalence and economic impact. It is therefore of great importance to understand the mechanisms of diabetes and to work toward the discovery of improved clinical treatments. These two objectives are also the ultimate goal of the study described in this dissertation. Toward this end, a series of experiments were designed to evaluate the ability of Ag-THC, a cannabinoid compound, to attenuate the MLDSTZ-induced autoimmune diabetes. Results obtained from these studies not only provided insight into the potential role of environmental Chemicals in the triggering of type I diabetes, but also contribute to a better understanding of the pharmacological and toxicological profiles of cannabinoid compounds. In the current investigation the effect of AQ-THC on MLDSTZ-induced diabetes was first examined in CD-1 mice, which is a mouse model of autoimmune diabetes previously established by Like and Rossini. The results from the present studies demonstrated a partial and transient protective effect by Ag-THC on the development of diabetes in this model. It was apparent from the cytokine profile that Ag-THC Inhibited a TH1-mediated immune response thereby attenuating pancreatic beta-cell damage initiated by MLDSTZ treatment. However, In spite of these data, It remains unclear whether Ag-THC would be capable of attenuating the development of diabetes in the spontaneous diabetes animal model and human Type I diabetes patients. As mentioned earlier, the MLDSTZ-Induced diabetes model represents a diabetic condition in which the 121 primary etiopathologic effect is produced by an environmental chemical. In this regard, etiopathogenesis in the MLDSTZ model is different from that underlying the disease in NOD mice, and BB rats which may involve a genetically programmed loss of tolerance to beta-cell specific antigens. Evidently, there is no single cause of type I diabetes in man, with disease induction representing a genetic susceptibility interacting with environmental triggers, such as toxins in the diet as well as pathogenic viruses. In order to clarify whether the protection produced by AQ-THC Is unique to this chemical-initiated diabetes model, future studies will be necessary to determine whether Ag-THC or other cannabinoid compounds can attenuate the disease process in animal models that develop diabetes spontaneously. However, to manipulate the timescale and the incidence of the diabetes development in those spontaneous models Is much more challenging, therefore the MLDSTZ model was employed here in these initial studies aimed at evaluating the protective effect of AQ-THC in a reproducible model of chemical-induced autoimmune diabetes. The present study further examined the effect of Ag-THC in MLDSTZ- induced diabetes in 86C3F1 mice for comparative purposes. The B603F1 mouse strain is a commonly used animal model in toxicology studies and was therefore employed in the present study to provide additional txicology-related information concerning genetic bases for susceptibility and resistance to diabetogenic environmental toxins. In contrast to CD-1 mice, BBC3F1 mice exhibited a less pronounced hyperglycemia and the absence of insulitis after MLDSTZ treatment and the protection by A9-THC was also less extensive. This 122 observation is consistent with the probability that MLDSTZ treatment Initiated direct Irreversible beta-cell damage in certain animal strains, even in the absence of a robust Immune response. These data supported previously reported results that different animal species and strains exhibit a markedly different sensitivity to MLDSTZ treatment In eliciting diabetogenic response. Accordingly it indicated that MLDSTZ-induced diabetes is a heterogeneous disorder which Is dependent on genetic factors as well as environmental factors. It Is intriguing that MLDSTZ-treated B6C3F1 mice exhibited an absence of insulitis as well as minimal protection by AQ-THC co-treatment in developing the diabetic phenotype. This is in contrast to CD-1 mice which exhibited strong insulitis and significantly greater protection by A9-THC. These results lead to two speculations. The first being that MLDSTZ-induced diabetes likely consists of two simultaneously mechanisms: direct cytotoxic action of STZ to Insulin- producing cells which is coupled with the induction of an immune response against insulin-producing cells. It was further suggested that the protective action of Ag-THC in STZ-induced diabetes was through the inhibition of the autoimmune response directed against the Insulin-producing cells. As a mattter of fact, In the case of chemical-induced immune response such as In the model of MLDSTZ- induced diabetes, It is difficult to separate the functional component of cell damage or cell loss attributed to the direct cytotoxicity of the toxin from the extent of functional impairment produced by cell-mediated immunity. In order to distinguish which of the two putative mechanisms (eg. direct alteration of beta- Cells and immune response) may be modulated by Ag-THC treatment, studies 123 were designed to examine the effect of Ag-THC treatment post STZ administration in CD-1 mice. Surprisingly, these studies demonstrated that A9- THC treatment was protective even when cannabinoid treatment was not initiated until the STZ treatment regimen was concluded. It is important to emphasized that the actions of STZ are extremely rapid in light of the fact that the half life of this compound in an aqueous environment is approximately 15 min. Therefore the protective effects produced by A9-THC treatment beginning on Day 6 are not attributable to an Inhibition of STZ cell uptake, altered metabolism or disruption of Its alkylating properties. Combined with the results observed in B6C3F1 mice, these results further support the contention that both the direct cytotoxic action of STZ and the Immune-based elements contributed to the diabetogenic effects of MLDSTZ treatment. Overall, these findings add to our current knowledge in assigning relative importance to the components participating in the ultimate beta-cell destruction in the MLDSTZ model. Furthermore, these studies contribute to our understanding of the mechanism by which environmental toxins elicits immune-mediated responses. In addition to the immunosuppressive effect produced by A9-THC, another contributing factor considered was a direct stimulatory effect of A9-THC on pancreatic insulin-producing cells. In agreement with a previous report by Laychock et al. using Isolated rat pancreatic islets (Laychock et al., 1986), the present study showed that Ag-THC stimulated Insulin release from both CD-1 mouse islets and rat derived insulin-producing cell line. Although It is well known 124 that A9-THC produces a variety of biological effects on different cell types and organs systems, there was relatively little information ayailable regarding the effects of cannabinoids on the pancreas, particularly on insulin-producing cells. For example, before our study, it was unclear whether there Is an expression of cannbinoid receptors in the pancreas; whether A9-THC could alter another important insulin-producing cell function, insulin synthesis; and if so, what were the intracellular signaling pathways involved in Ag-THC-induced insulin release. As an initial attempt to facilitate a better understanding of this undeveloped area, important signaling pathways were characterized In the current investigation to provide additional information on the mechanism of the A9-THC-stimulated insulin release observed in this study. It was suggested from the data obtained here that Ag-THC-stimulated insulin release is mediated through an elevation of intracellular calcium through the opening of calcium channels via a CB1- dependent mechanism. These findings expand present knowledge and may stimulate future studies concerning the pharmacological and toxicological profile of cannabinoid compounds. However, it is important to note that results from our in vivo studies presented in Chapter II showed that A9-THC treatment alone did not alter serum glucose and pancreatic insulin in CD-1 mice not treated with MLDSTZ. An insulin-releasing effect of A9-THC on mice treated the cannabinoid alone would be expected to produce hypoglycemia and possibly a reduction in pancreatic insulin. Several considerations should be made when analyzing these results 125 obtained in vivo. First, when possible A°-THC-Induced hypoglycemia was examined in vivo in the present investigation, non-fasting serum glucose concentrations were measured In the experimental animals. This measurement is valid to detect a possible diabetic state, but within the normal physiological range, blood glucose can vary markedly in non-fasted animals. Therefore, to help assess a more precise picture of a possible insulin stimulatory effect of A9- THC occurring in vivo, a thorough analysis of glucose and insulin levels, such as a glucose tolerance test (GTT) after Ag-THC treatment, should be conducted In future studies. Second, it is possible that only hyperglycemic animals would respond to Ag-THC treatment-induced insulin release under a very high glucose condition. This could explain why control mice treated with A9-THC alone did not exhibit elevated pancreatic Insulin or reduced blood glucose. However, this interpretation is countered by a previous observation (Laychock et al., 1986) and the results obtained from the current study, showing an insulinotropic effect of A9- THC at lower (e.g. fasting) glucose levels. It should be recognized that a biological effect in vivo Involves a more complex system of insulin release and blood glucose regulation. For example, in non-diabetic normal animals, where the net result of hormonal regulation is capable of maintaining a normal hormone balance, a Ag-THC-induced insulin release may be counteracted by actions of glucagon and somatostatin. Hence, the blood glucose concentration measured in normal CD-1 mice may remain within normal physiological limits upon Ag-THC treatment. Third, it is possible that the effect of A9-THC on pancreatic beta-cell 126 functions may only involve a stimulation of insulin release with no modulation of insulin synthesis. If insulin synthesis was not also modulated by AQ-THC, the total insulin content in the pancreas may remain unchanged after treatment with A9- THC. Therefore, It Is not surprising that the pancreatic insulin content measured in CD-1 mice in our in vivo study was not altered by Ag-THC treatment, neither was the blood glucose concentration. In conclusion, our results demonstrate that Ag-THC treatment attenuated the hyperglycemia produced by MLDSTZ treatment in CD-1 mice. Moreover, the attenuation of MLDSTZ-induced hyperglycemia was partly due to the inhibition of the immune response by THC directed against the pancreatic Islets. In addition to the Immune suppression, our studies also suggest that Ag-THC exerts direct effects on insulin-producing cells by directly stimulating the release of Insulin. Stimulation of insulin release by Ag-THC is mediated through a CB1 dependent mechanisms which involves the rapid rise of [Ca”], through verapamil-sensitive channels and the activation of CaM kinase II. In spite of these data, it is difficult to determine whether the protective effect of Ag-THC on MLDSTZ-induced diabetes is due to modulation of the immune system and/or due to direct alteration of beta-cell function since these two mechanisms are not easily separated in vivo. In order to further ascertain the relative contribution of each of these two mechanisms other approaches will be necessary. For example, in vitro studies to further examine the effect of Ag-THC on insulin release and insulin synthesis In MLDSTZ-treated mouse islets may present a direction for the future 127 investigations. Furthermore, the CD-1 scid\scid mouse currently being developed at Charles River Laboratory would also be useful for future studies to test the intervention protocol with A9-THC in the MLDSTZ model, because it lacks the immune component. 128 BIBLIOGRAPHY 129 BIBLIOGRAPHY AGARWAL, M.: Streptozotocin: Mechanisms of action. FEBS.Lett. 120: 1-3, 1980. AIZAWA, T., YADA, T., ASANUMA, N., SATO, Y., ISHIHARA, F., HAMAKAWA, N., YAEKURA, K. AND HASHIZUME, K.: Effects of thapsigargin, an intracellular CA2+ pump inhibitor, on insulin release by rat pancreatic B- cell. Life.Sci. 57: 1375-1381, 1995. ASHCROFT, S.J. AND HUGHES, S.J.: Protein phosphorylation In the regulation of Insulin secretion and biosynthesis. Biochem.Soc.Trans. 18: 116-118, 1990. BANNISTER, B.: The synthesis and biological activities of some analogs of streptozotocin. J. Antibotics. 25: 377-385, 1972. BARTH, F. AND RINALDI, C.M.: The development of cannabinoid antagonists. Curr.Med.Chem. 6: 745-755, 1999. BEARDSLEY, P.M. AND MARTIN, B.R.: Effects of the cannabinoid CB(1) receptor antagonist, SR141716A, after Delta(9)—tetrahydrocannabinol withdrawal. Eur.J. Pharmacol. 387: 47-53, 2000. BEATTIE, G., LANNOM, R., LIPSICK, J., KAPLAN, N. AND OSLER, A.: Streptozotocin-induced diabetes In athymic and conventional BALB/c mice. Diabetes 29: 146-150, 1980. BERNARDIS, L.L. AND FROHMAN, L.A.: Plasma growth hormone responses to electrical stimulation of the hypothalamus in the rat. Neuroendocrinology. 7: 193-201, 1971. BLANCHARD, D.K., DJEU, J.Y., KLEIN, T.W., FRIEDMAN, H. AND STEWART, W.E.: Interferon-gamma induction by Iipopolysaccharide: dependence on Interleukin 2 and macrophages. J.|mmunol. 136: 963-970, 1986. BLUE, M. AND SHIN, 8.: Diabetes induction by subdiabetogenic doses of streptozotocin in BALB/CBOM mice. Non-involvement of host B- lymphocyte functions. Diabetes 33: 105-110, 1984. BOLAFFI, J., NAGAMATSU, S. AND HARRIS, J.e.al.: Protection by thymidine, an inhibitor of polyadenosine diphosphate ribosylation, of streptozotocin inhibition of Insulin secretion. Endocrinology 120: 21171987. BOLAFFI, J., NOWLAN, R., CRUZ, L. AND GRODSKY, G.: Progressive damage of cultured pancreatic islets after single early exposure to streptozotocin. 130 Diabetes 35: 1027-1033, 1986. BONNER-WEIR, S. AND SMITH, F.E.: Joslin's Diabetes, pp. 15-16, Philadelphia, PA, 1994. BOUABOULA, M., POINOT, C.C., MARCHAND, J., CANAT, X., BOURRIE, B., RINALDI, C.M., CALANDRA, B., LE-FUR, G. AND CASELLAS, P.: Signaling pathway associated with stimulation of C82 peripheral cannabinoid receptor. Involvement of both mitogen-activated protein kinase and Induction of Krox-24 expression. Eur.J.Biochem. 237: 704- 711, 1996. BOUABOULA, M., RINALDI, M., CARAYON, P., CARILLON, C., DELPECH, B., SHIRE, 0., LE FUR, G. AND CASELLAS, P.: Cannabinoid-receptor expression In human leucytes. Eur.J. Biochem. 214: 173-180, 1993. BRAIMAN, L., SHEFFI, F.L., BAK, A., TENNENBAUM, T. AND SAMPSON, S.R.: Tyrosine phosphorylation of specific protein kinase C isoenzymes participates in insulin stimulation of glucose transport In primary cultures of rat skeletal muscle. Diabetes 48: 1922-1929, 1999. BROWN, J.L.: N-Nitrosamines. Occup.Med. 14: 839-848, 1999. CABRAL, G., LOCKMULLER, J. AND MISHKIN, E.: A9-tetrahydrocannabinol decreases serum alpha/beta interferon response to heeerpes simplex virus type 2 in the b6C3F1 mouse. Proc.Soc.Exp.Biol.Med. 181: 305-311, 1986b. CABRAL, G., MISHKIN, E., MARCIANO-CABRAL, F., COLEMAN, P., HARRIS, L. AND MUNSON, A.: Effect of A9—tetrahydrocannabinol on herpes simplex virus type2 vaginal Infection in the guinea pig. Proc.Soc.Exp.Biol. Med. 182: 181-186, 1986a. CAHILL, G. AND MCDEVITT, H.: Insulin-dependent diabetes: the initial lesion. N.Ecgl.J.Med. 304: 1454-1463, 1981. CAMPBELL, |., OXBROW, L., KOULMANDA, M. AND HARRISON, L.: IFN- gamma induces islet cell MHC antigens and enhances autoimmune, streptozotocin-induced diabetes in the mousse. J Immunol 140: 1111- 1116, 1988. CAMPBELL, I., WONG, G., SCHRADER, J. AND HARRISON, L.: Interferon- gamma enhances the expression of the major hiscompatibility class I antigens on mouse pancreatic B cells. Diabetes 34 : 1205-1209, 1985. CARCHMAN, R.A., END, D.W., DEWEY, W.L. AND WARNER, W.: Marihuana and opiate interactions with hormonal induced systems in cell culture. 131 Prog.Clin.Biol.Res. 27: 237-252, 1979. CARTER, J., CARLSON, J., FOWLER, J., HARTENBACH, E., ADCOCK, L., CARSON, L. AND TWIGGS, L.B.: Invasive vulvar tumors in young women-—a disease of the immunosuppressed? Gynecol.Oncol. 51: 307- 310, 1993. CAULFIELD, M.P. AND BROWN, D.A.: Cannabinoid receptor agonists inhibit Ca current in NG108-15 neuroblastoma cells via a pertussis toxin-sensitive mechanism. Br.J Pharmacol. 106: 231-232, 1992. CHAIT, L.D. AND ZACNY, J.P.: Reinforcing and subjective effects of oral delta 9-THC and smoked marijuana in humans. Psychopharrnacology Berl. 107: 255-262, 1992. CHAPERON, F. AND THIEBOT, M.H.: Behavioral effects of cannabinoid agents in animals. Crit.Rev.Neurobiol. 13: 243-281, 1999. COHEN, D.: Magnetoencephalography: detection of the brain's electrical activity with a superconducting magnetometer. Science 175: 664-666, 1972. COLOMBO, G., AGABIO, R., DIAZ, G., LOBINA, C., REALI, R. AND GESSA, G.L.: Appetite suppression and weight loss after the cannabinoid antagonist SR 141716. Life Sci. 63: L113-L1171998. CONDIE, R., HERRING, A., LEE, M., KOH, W. AND KAMINSKI, N.: Cannabinoid inhibition of adenylate cyclase-mediated signal transduction and interleukin 2 (IL-2) expression in the murine T-cell line, EL4.lL-2. J Biol Chem 271: 13175-13183, 1996. COUTTS, A.A., BREWSTER, N., INGRAM, T., RAZDAN, R.K. AND PERTWEE, R.G.: Comparison of novel cannabinoid partial agonists and SR141716A in the guinea-pig small intestine. Br.J Pharmacol. 129: 645-652, 2000. CRAIGHEAD, J.: Current views of insulin-dependent diabetes mellitus. N Engl J Med 299: 1439-1445, 1978. CUI, L., YU, W.P. AND PALLEN, C.J.: Insulin secretagogues activate the secretory granule receptor-like protein-tyrosine phosphatase IAR. J.Biol.Chem. 273: 34784-34791, 1998. DALTERIO, S., BARTKE, A., BLUM, K. AND SWEENEY, C.: Marihuana and alcohol: perinatal effects on development of male reproductive functions in mice. Prog.Biochem.Pharmacol. 18: 143—154, 1981. DAS, S.K., PARIA, B.C., CHAKRABORTY, I. AND DEY, S.K.: Cannabinoid ligand-receptor signaling in the mouse uterus. Proc.Natl.Acad.Sci.U.S.A. 132 92: 4332-4336, 1995. DEAN, P.M. AND MATTHEWS, E.K.: Electrical activity in pancreatic islet cells. Nature 219: 389-390, 1968. DEAN, P.M. AND MATTHEWS, E.K.: Electrical activity in pancreatic islet cells: effect of ions. J.Physiol.Lond. 210: 265-275, 1970. DEVASKAR, S.U., SINGH, B.S., CARNAGHI, L.R., RAJAKUMAR, P.A. AND GIDDINGS, S.J.: Insulin II gene expression in rat central nervous system. Regul.Pept. 48: 55-63, 1993. DOW, G.J. AND MEYERS, F.H.: The California program for the investigational use of THC and marihuana in heterogeneous populations experiencing nausea and vomiting from anticancer therapy. J Clin.PharmacoI. 21: 1288-132S, 1981. EASOM, R.A.: CaM kinase II: a protein kinase with extraordinary talents germane to Insulin exocytosis. Diabetes 48: 675-684, 1999. EMDIN, S.O., DODSON, G.G., CUTFIELD, J.M. AND CUTFIELD, S.M.: Role of zinc in insulin biosynthesis. Some possible zinc-insulin interactions in the pancreatic B-Cell. Diabetologia 19: 174-182, 1980. ENDO, T. AND HIDAKA, H.: Ca2+-calmodulin dependent phosphorylation of myelin isolated from rabbit brain. Biochem.Biophys.Res.Commun. 97: 553-558, 1980. FAJEN, J.M., CARSON, G.A., ROUNBEHLER, D.P., FAN, T.Y., VITA, R., GOFF, U.E., WOLF, M.H., EDWARDS, G.S., FINE, D.H., REINHOLD, V. AND BIEMANN, K.: N-nitrosamines in the rubber and tire industry. Science 205: 1262-1264, 1979. FARR, A., MANNSCHRECK, S. AND ANDERSON, 8.: Expression of class II MHC antigens in murine pancreas after streptozotocin-induced insulitis. Diabetes 37: 1373-1379, 1988. FELDER, C.C. AND GLASS, M.: Cannabinoid receptors and their endogenous agonists. Annu.Rev.Pharmacol.Toxicol. 38: 179-200, 1998. FELDER, C.C. AND GLASS, M.: Cannabinoid receptors and their endogenous agonists. Annu.Rev.Pharmacol.Toxicol. 38: 179-200, 1998. FELDER, C.C., JOYCE, K.E., BRILEY, E.M., GLASS, M., MACKIE, K.P.. FAHEY, K.J., CULLINAN, G.J., HUNDEN, D.C., JOHNSON, D.W., CHANEY, M.O., KOPPEL, G.A. AND BROWNSTEIN, M.: LY320135, a novel cannabinoid CB1 receptor antagonist, unmasks coupling of the CB1 receptor to stimulation of cAMP,accumulation. J.Pharmacol.Exp.Ther. 133 284: 291 -297, 1998. FELDER, C.C., JOYCE, K.E., BRILEY, E.M., MANSOURI, J., MACKIE, K., BLOND, O., LAI, Y., MA, A.L. AND MITCHELL, R.L.: Comparison of the pharmacology and signal transduction of the human cannabinoid CB1 and CB2 receptors. Mol.Pharmacol. 48: 443-450, 1995. FELDER, C.C., VELUZ, J.S., WILLIAMS, H.L., BRILEY, E.M. AND MATSUDA, L.A.: Cannabinoid agonists stimulate both receptor- and non-receptor- mediated signal transduction pathways in cells transfected with and expressing cannabinoid receptor clones Mol.Pharmacol. 42: 838-845, 1992. FELDER, C., JOYCE, K., BRILEY, E., MANSOURI, J., MACKIE, K., BLOND, O., LAI, Y., MA, A. AND MITCHELL, R.: Comparison of the pharmacology and signal transduction of the human cannabinoid CB1 and CB2 receptors. Mol Pharmacol 48: 443-450, 1995. FILIPEANU, C.M., DE-ZEEUW, D. AND NELEMANS, S.A.: Delta-9- tetrahydrocannabinol activates [Ca2+]i Increases partly sensitive to capacitative store refilling. Eur.J.Pharmacol. 336: R1 -R3,1 997. FINDLAY, I. AND DUNNE, M.J.: Voltage-activated Ca2+ currents in Insulin- secreting cells. FEBS Lett. 189: 281-285, 1985. FlSCHER-STENGER, K., DOVE PETITIT, D. AND CABRAL, G.: A9- tetrahydrocannabinol inhibiiton of tumor necrosis factor-a: suppression of post-translational events. J. Pharmacol.Exp.Ther. 267: 1558-1565, 1993. FISCHER, L.J., WAGNER, M.A. AND MADHUKAR, B.V.: Potential involvement of calcium, CaM kinase II, and MAP kinases in PCB-stimulated insulin release from RINm5F cells. Toxicol.Appl.Pharmacol. 159: 194-203, 1999 FISCHER, L.J.: Toxicity to the insulin-secreting beta-cell. In Comprehensive Toxicology, ed. by l. Glenn Sipes, A. McQueen and A. Jay Gandolfi, pp. 701-724, Pergamon, New York, NY, 1997. FISCHER, L., WOLD, J. AND RICKERT, D.: Structure activity relationships in drug-induced pancreatic islet cell toxicity. Toxicol Appl Pharrncol 26: 288- 298, 1973. GANDA, O.P., ROSSINI, A.A. AND LIKE, A.A.: Studies on streptozotocin diabetes. Diabetes 25: 595-603, 1976. GARCIA, B.M., REVERTE, M. AND MORATINOS, J.: Role of Ca2+ channel blockers in Insulin secretion resulting from alpha 1- and beta-adrenoceptor stimulation in the rabbit. Eur.J.Pharmacol. 219: 461-464, 1992. 134 GEBREMEDHIN, D., LANGE, A.R., CAMPBELL, W.B., HILLARD, C.J. AND HARDER, D.R.: Cannabinoid CB1 receptor of cat cerebral arterial muscle functions to Inhibit L-type Ca2+ channel current. Am.J.Physiol. 276: H2085-H20931999. GEPTS, W.: Pathological anatomy of the pancreas in juvenile diabetes mellitus. Diabetes 14: 619-633, 1965. GERARD, C., MOLLEREAU, G., VASSART, G. AND PARMENTIER, M.: Nucleotide sequence of a human cannabinoid receptor cDNA. Nucleic Acids res 18: 71421990. GERARD, C., MOLLEREAU, G., VASSART, G. AND PARMENTIER, M.: Molecular cloning of a human cannabinoid receptor which is also expressed in testis. Biochem J 279: 129-134, 1991. GIDDINGS, S.J., CARNAGHI, L.R., FISCHER, L.J. AND MILLER, C.P.: Differential regulation of rat insulin I and II messenger RNA synthesis: effects of fasting and cyproheptadine. Mol.Endocrinol. 5: 549-554, 1991. GIDDINGS, S.J., CHIRGWIN, J.M. AND PERMU'IT, M.A.: Glucose regulated insulin biosynthesis in isolated rat pancreatic islets is accompanied by changes in proinsulin mRNA. Diabetes Res. 2: 71-75, 1985. GLASS, M. AND DRAGUNOW, M.: Induction of the Krox 24 transcription factor in striosomes by a cannabinoid agonist. Neuroreport. 6: 241-244, 1995. GLASS, M. AND NORTHUP, J.K.: Agonist selective regulation of G proteins by cannabinoid CB(1) and CB(2) receptors. Mol.Pharmacol. 56: 1362-1369, 1999. GONG, H.J., TASHKIN, D., SIMMONS, M., CALVARESE, B. AND SHAPIRO, B.: ‘ Acute and subacute bronchial effects of oral cannabinoids. Clin Pharmacol Ther 35: 26-32, 1984. GORTER, R.W.: Cancer cachexia and cannabinoids. Forsch. Komplementarmed. 6 Suppl 3: 21-22, 1999. GREGERSEN, S., THOMSEN, J.L. AND HERMANSEN, K.: Endothelin-1 (ET-1)- potentiated insulin secretion: involvement of protein kinase C and the ET(A) receptor subtype. Metabolism 49: 264-269, 2000. GROMADA, J., HOY, M., RENSTROM, E., BOKVIST, K., ELIASSON, L., GOPEL, S. AND RORSMAN, P.: CaM kinase lI-dependent mobilization of secretory granules underlies acetylcholine-induced stimulation of exocytosis in mouse pancreatic B-0ells. J Physiol.Lond. 518: 745-759, 1999. 135 GROMADA, J., DING, W.G., BARG, S., RENSTROM, E. AND RORSMAN, P.: Multisite regulation of insulin secretion by CAMP-increasing agonists: evidence that glucagon-like peptide 1 and glucagon act via distinct receptors. Pflugers Arch. 434: 515-524, 1997. HAHN, H.J., HAGEN, R., KNOBLOCH, C. AND BESCH, W.: Plasma glucose and pancreatic insulin content in the early phase of streptozotocin-induced diabetes (preliminary results). Acta Biol.Med.Ger. 40: 83-85, 1981. HALBAN, P.A., PRAZ, G.A. AND WOLLHEIM, C.B.: Abnormal glucose metabolism accompanies failure of glucose to stimulate insulin release from a rat pancreatic cell line (RINm5F). Biochem.J 212: 439-443, 1983. HAMMONDS, P., SCHOFIELD, P.N. AND ASHCROFT, S.J.: Glucose regulates preproinsulin messenger RNA levels in a clonal cell line of simian virus 40-transformed B cells. FEBS Lett. 213: 149-154, 1987. HARRIS, L.S., CARCHMAN, R.A. AND MARTIN, B.R.: Evidence for the existence of specific cannabinoid binding sites. Life Sci. 22: 1131-1137, 1978. HATOUM, N.S., DAVIS, W.M., ELSOHLY, M.A. AND TURNER, C.E.: Perinatal exposure to cannabichromene and delta 9-tetrahydrocannabinol: separate and combined effects on viability of pups and on male reproductive system at maturity. Toxicol.Lett. 8: 141-146, 1981. I-IAVERY, 0.0. AND FAZIO, T.: Estimation of volatile N-nitrosamines In rubber nipples for babies‘ bottles. Food Chem.Toxicol. 20 : 939-944, 1982. HENQUIN J.C.: Cell biology and Insulin secretion. In Joslin's diabetes mellitus, ed. by Kahn CR. and Weir G.C., pp. 66-68, lea & Febiger, Malvem, PA, 1994. . HO, B.Y., UEZONO, Y., TAKADA, S., TAKASE, I. AND IZUMI, F.: Coupling of the expressed cannabinoid CB1 and CBZ receptors to phospholipase C and G protein-coupled inwardly rectifying K+ channels. Receptors.Channels. 6: 363-374, 1999. HOLDCROFT, A., SMITH, M., JACKLIN, A., HODGSON, H., SMITH, B., NEWTON, M. AND EVANS, F.: Pain relief with oral cannabinoids in familial Mediterranean fever. Anaesthesia 52: 483-486, 1997. HOLZ, G.G., LEECH, C.A., HELLER, R.S., CASTONGUAY, M. AND HABENER, J.F.: CAMP-dependent mobilization of intracellular Ca2+ stores by activation of ryanodine receptors in pancreatic beta-cells. A Ca2+ signaling system stimulated by the insulinotropic hormone glucagon-like peptide-1-(7-37). J.Biol.Chem. 274: 14147-14156, 1999. 136 HOWELL, S.L., JONES, P.M. AND PERSAUD, S.J.: Protein kinase C and the regulation of Insulin secretion. Biochem.Soc.Trans. 18: 114-116, 1990. HOWELL, S.L. AND TYHURST, M.: Microtubules, microfilaments and insulin- secretion. Diabetologia 22: 301 -308, 1982. HOWLET‘I’, AC: The CB1 cannabinoid receptor In the brain. Neurobiol.DIs. 5: 405-416, 1998. HUANG, S. AND TAYLOR, G.: immune insulitis and antibodies to nucleic acids Induced with streptozotocin in mice. Clin Exp Immunol 43: 425-429, 1981. HUGHES, S.J., CHALK, J.G. AND ASHCROFT, S.J.: The role of cytosolic free . Ca2+ and protein kinase C in acetylcholine-Induced Insulin release in the clonal beta-cell line, HIT-T15. Biochem.J. 267: 227-232, 1990. HUNTER, S.A. AND BURSTEIN, S.H.: Receptor mediation in cannabinoid stimulated arachidonic acid mobilization and anandamide synthesis. Life Sci. 60: 1563-1573, 1997. ILKOVA, H.M.: Phosphoinositide metabolism and insulin secretion. Diabete.Metab. 20: 150-156, 1994. ITOH, N., SEI, T., NOSE, K. AND OKAMOTO, H.: Glucose stimulation of the proinsulin synthesis in isolated pancreatic islets without increasing amount of proinsulin mRNA. FEBS Lett. 93: 343-347, 1978. IWAKIRI, R., NAGAFUCHI, S., KOUNOUE, E., NAKANO, S., KOGA, T., NAKAYAMA, M., NAKAMURA, M. AND NIHO, Y.: Enhancement of streptozotocin (STZ) induced diabetes by cyclosporin A (CYA) in mice. Diabetes 35 (Suppl 1): 183A1986. JARAI, Z., WAGNER, J.A., VARGA, K., LAKE, K.D., COMPTON, D.R., MARTIN, B.R., ZIMMER, A.M., BONNER, T.I., BUCKLEY, N.E., MEZEY, E.. RAZDAN, R.K., ZIMMER, A. AND KUNOS, G.: Cannabinoid-induced mesenteric vasodilation through an endothelial site distinct from CB1 or CB2 receptors. Proc.Natl.Acad.Sci.U.S.A. 96: 14136-14141, 1999. JARRETT, R.J., KEEN, H. AND TRACK, N.: Glucose and RNA synthesis in mammalian Islets of Langerhans. Nature 213: 634-635, 1967. JEON, Y., YANG, K., PULASKI, J. AND KAMINSKI, N.: Attenuation of iNOS gene expression by A 9-tetrahydrocannabinol is mediated through the inhibition of NF-kB/rel activation. Mol Pharm 50: 334-341, 1996. KAMINSKI, N.E.: Inhibition of the CAMP signaling cascade via cannabinoid receptors: a putative mechanism of immune modulation by cannabinoid 137 compounds. Toxicol.Lett. 102-103: 59-63, 1998. KAMINSKI, N.E., ABOOD, M.E., KESSLER, F.K., MARTIN, B.R. AND SCHATZ, A.R.: Identification of a functionally relevant cannabinoid receptor on mouse spleen cells that is involved in cannabinoid-mediated immune modulation. Mol.Pharmacol. 42: 736-742, 1992. KAMINSKI, N.E., KOH, W.S., YANG, K.H., LEE, M. AND KESSLER, F.K.: Suppression of the humoral immune response by cannabinoids Is partially mediated through inhibition of adenylate cyclase by a pertussis toxin- sensitive G-protein coupled mechanism. Biochem.PharmacoI. 48: 1899- 1908, 1994. KAMINSKI, N., ABOOD, M., KESSELER, F., MARTIN, B. AND SCHARTZ, A.: Identification of a functionally relevant cannabinoid receptor on mouse spleen cells involved in cannabinoid-mediated immune modulation. Mol Pharmacol 42: 736-742, 1992. KAMINSKI, N., KOH, W., LEE, M., YANG, K. AND KESSELER, F.: Suppression of the humoral immune response by cannabinoids is partially mediated through inhibition of adenylate cyclase by a pertussis toxin-sensitive G- proytein coupled mechanism. Biochem Pharmacol 48: 1899-1908, 1994. KARAM, J., LEWITT, P., YOUNG, C., NOWLAIN, R., FRANKEL, B., FUJIYAA, H., FREEDMAN, Z. AND GRODSKY, G.: lnsulinopenic diabetes after rodenticide (Vacor) ingestion: a unique model of acquired diabetes in man. Diabetes 29: 971-978, 1980. KATZ, D., BENOIST, C. AND MATHIS, D.: T helper cell subsets in insulin- dependent diabetes. Science 268: 1 185-1 188, 1995. KAWADA, J., TOIDE, K., NISHIDA, M., YOSHIMURA, Y. AND TSUJIHARA, K.: New diabtogenic streptozotcin analog,3-O-methyl-2-(methylnitrosoamino)- carbonyl amino-D-glucopyranosezevidence for a glucose recognitionsite on pancreatic B-Cells. Diabetes 35: 162-163, 1986. KAWAKAMI, Y., KLEIN, T.W., NEWTON, C., DJEU, J.Y., SPECTER, S. AND FRIEDMAN, H.: Suppression by delta-9-tetrahydrocannabinol of interleukin 2-induced lymphocyte proliferation and lymphokine-activated killer cell activity. lnt.J lmmunopharmacol. 10: 485-488, 1988. KIESEL, U., FALKENBERG, F. AND KOLB, H.: Genetic control of low-dose streptozotcin-induced autoimmune diabetes In mice. J Immunol 130: 1719-1722, 1983. KIESEL, U. AND KOLB, H.: Low-dose streptozotocin-induced autoimmune diabetes Is under the genetic control of the major histocompatibility 138 complex In mice. Diabetologia 23: 69-71, 1982. KIESEL, U., OSCHILEWSKI, M., TANIGUCHI, M. AND KOLB, H.: Modulation of low-dose streptozotocin-induced diabetes in mice by administration of antibodies to l-A, l-E and l-J determinants. Diabetologia 32: 173—176, 1989. KILHOFFER, M.C., LUKAS, T.J., WATTERSON, D.M. AND HAIECH, J.: The heterodimer calmodulin: myosin light-chain kinase as a prototype vertebrate calcium signal transduction complex. Biochim.Biophys.Acta 1160: 8-15, 1992. KISHI, H., YE, L.H., NAKAMURA, A., OKAGAKI, T., IWATA, A., TANAKA, T. AND KOHAMA, K.: Structure and function of smooth muscle myosin light chain kinase. Adv.Exp.Med.BioI. 453: 229-234, 1998. KLEIN, T.W., NEWTON, C. AND FRIEDMAN, H.: Cannabinoid receptors and immunity. lmmunol.Today19: 373-381, 1998. KLINKHAMMER, C., DOHLE, C. AND GLEICHMANN, H.: T cell-dependent Class II major histocompatibility complex antigen expression in vivo induced by the diabetogen streptozotocin. Immunobiology 180(1): 1-11, 1989. KLINKHAMMER, C., POPOWA, P. AND GLEICHMANN, H.: Specific immunity to the diabetogen streptozotocin: celllular requirements for induction of Iymphoproliferation. Diabtes 37: 74-80, 1988. KOLB, H.: Mouse models of insulin dependent diabetes: low-dose streptozotocin-induced diabetes and nonobese diabetic (NOD) mice. Diabetes Metab Rev 3: 751-778, 1987. KOLB, H., OSCHILEWSKI, M., SCHWAB, E., OSCHILEWSKI, U. AND KIESEL, U.: Effect of cyclosporin A on low-dose streptozotocin diabetes in mice. Diabetes Res 2: 191-193, 1985. KOSTOWSKI, W. AND TARCHALSKA, B.: The effects of some drugs affecting brain 5-HT on the aggressive behaviour and spontaneous electrical activity of the central nervous system of the ant, Formica rufa. Brain Res. 38: 143-149, 1972. KROMANN, H., CHRISTY, M., EGEBERG, J., LERNMARK, A. AND NERUP, J.: The low dose streptozotocin murine model type I (Insulin-dependent) diabetes mellitus: studies in vivo and invItro of the modulating effects sex hormones. Diabetologia 2: 194-198, 1982. KRZYSTYNIAK, K., KOZLOWSKA, E., DESJARDINS, R., DRELA, N., KOWALCZYK, R., KARWOWSKA, K. AND IZDEBSKA-SZYMONA, K.: Different T-Cell activation by streptozotocin and Freund's adjuvant in 139 popliteal lymph node (PLN). Int J lmmunophannacol 17: 189-196, 1995. KUTTLER, B. AND SCHNEIDER, E.: Diabetes mellitus in mice induced by multiple subdiabetogenic doses of streptozotocin: age and sex dependence. Acta Biol Med Germ 41: 1199-1202, 1982. LANG, J., USHKARYOV, Y., GRASSO, A. AND WOLLHEIM, C.B.: Ca2+- independent Insulin exocytosis induced by alpha-Iatrotoxin requires latrophilin, a G protein-coupled receptor. EMBO J. 17: 648-657, 1998. LANGE, K. AND BRANDT, U.: The lP3-sensitive calcium store of HIT cells Is located in a surface-derived vesicle fraction. FEBS Lett. 320: 183-188, 1993. LAY, L., ANGUS, J.A. AND WRIGHT, C.E.: Pharmacological characterisation of cannabinoid CB(1) receptors in the rat and mouse. Eur.J Pharmacol. 391: 151-161, 2000. LAYCHOCK, S.G.: Rat pancreatic islet and RINm5F cell responses to epiandrosterone, dehydroepiandrosterone and interleukin-1 beta. Biochem.Pharmacol. 55: 1453-1464, 1998. LAYCHOCK, S., HOFFMAN, J., MEISEL, E. AND BILGIN, S.: Pancreatic islet arachidonic acid turnover and metabolism and Insulin release in response to delta-9-tetrahydrocannabinol. Biochem-Pharmacol 35: 2003-2008, 1986. LAZARUS, 8.8. AND SHAPIRO, S.H.: Streptozotocin-induced diabetes and islet cell alterations in rabbits. Diabetes 21: 129-137, 1972. LE, P. AND LEITER, E.: Genetic control of diabetes induction by multiple low doses of streptozotocin in two BALB/c sublines. The Jackson Laboratory Annual reports 1986: 1-2, 1986. LE, P., LEITER, E. AND LEYENDECKER, J.: Genetic control of susceptibility to streptozotocin diabetes in inbred mice: effect of testosterone and H-2 haplotype. Endocrinology 116: 2450-2455, 1985. LEDOUX, S. AND WILSON, 6.: Effects of streptozotocin on a Clonal isolate of rat insulinoma cells. Biochim Biochim Biophys Acta 804: 387-392, 1984. LEDOUX, S., WOODLEY, S., PAITON, N. AND WILSON, G.: Mechanisms of nitrosamide-induced [3 cell damage: alterations in DNA. Daibetes 35: 866- 872, 1986. LEIERS, T., BIHLMAYER, A., AMMON, H.P. AND WAHL, M.A.: [Ca(2+)](I)- and insulin-stimulating effect of the non-membranepermeable phosphatase- 140 inhibitor microcystin-LR In intact insulin-secreting cells (RINm5F). Br.J Pharmacol. 130: 1406-1410, 2000. LEITER, E.: Multiple low-dose streptozotocin-induced hyperglycemia andinsulitis in 05781 mice: influence of inbred background,sex, and androgens. Proc Natl Acad Sci USA 79: 630-634, 1982. LEVENES, C., DANIEL, H., SOUBRIE, P. AND CREPEL, F.: Cannabinoids decrease excitatory synaptic transmission and impair long-term depression in rat cerebellar Purkinje cells. J Physiol.Lond. 510: 867-879, 1998. LIBLAU, R., SINGER, 8. AND MCDEVITT, H.: Th1 and Th2 CD4+ T cells In the pathogenesis of organ-specific autoimmune diseases. Immunol Today 16: 34-38, 1995. LIKE, A. AND ROSSINI, A.: Streptozotocin-induced pancreatic insulitis: new model of diabetes mellitus. Science 193: 1976. LITTLE, P. AND MARTIN, B.: The effects of delta 9-tetrahydrocannabinol and other cannabinoids on CAMP accumulation in synaptosomes. life Sci 48: 1133-1141,1991. LIU, K.S., WANG, C.Y., MILLS, N., GYVES, M. AND ILAN, J.: Insulin-related genes expressed in human placenta from normal and diabetic pregnancies. Proc.Natl.Acad.Sci.U.S.A. 82: 3868-3870, 1985. LU, Q., STRAIKER, A. AND MAGUIRE, G.: Expression of CB2 cannabinoid receptor mRNA In adult rat retina. Vis.Neurosci. 17: 91-95, 2000. MACLAREN, N. AND ALKINSON, M.: Insulin-dependent diabetes mellitus: the hypothesis of molecular mimicry between islet cell antigens and microorganisms. Mol Med Today 3: 76-83, 1997. MACLAREN, N., NEUFELD, M., MCLAUGHLIN, J. AND TAYLOR, G.: Androgen sensitization of streptozotocin-induced diabetes in mice. Diabetes 29: 710-716, 1980. MAECHLER, P., KENNEDY, E.D., SEBO, E., VALEVA, A., POZZAN, T. AND WOLLHEIM, C.B.: Secretagogues modulate the calcium concentration in the endoplasmic reticulum of Insulin-secreting cells. Studies in aequorin- expressing intact and permeabilized ins-1 cells. J Biol.Chem. 274: 12583- 12592, 1999. MAKINO, S. AND TOCHINO, Y.: The spontaneously non-obese-diabetic mouse. Exp Anim 27: 27-29, 1978. MALAISSE, W.J.: Insulin secretion and food intake. Proc.Nutr.SoC. 31: 213-217, 141 1972. MALAISSE, W.: The anomeric malaise: a manifestation of B-Cell glucotoxicity. Horm Metab Res 23: 307-311, 1991. MALLET, P.E. AND BENINGER, R.J.: The cannabinoid CB1 receptor antagonist SR141716A attenuates the memory Impairment produced by delta9- tetrahydrocannabinol or anandamide. Psychopharmacology Berl. 140: 11- 19, 1998. MANABE, S. AND WADA, O.: Triphenyltin fluoride (TPTF) as a diabetogenic agent. TPTF induces diabetic Iipemia by inhibiting insulin secretion from morphologically intact rabbit B—cells. Diabetes 30: 1013-1021, 1981. MASCIA, M.S., OBINU, M.C., LEDENT, C., PARMENTIER, M., BOHME, G.A., IMPERATO, A. AND FRATTA, W.: Lack of morphine-Induced dopamine release in the nucleus accumbens of cannabinoid CB(1) receptor knockout mice. Eur.J Pharmacol. 383: R1 -R21 999. MATSUDA, L., LOLAIT, S., BROWNSTEIN, M., YOUNG, A. AND BONNER, T.: Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 346: 561-564, 1990. MECHOULAM, R.: Marihuana chemistry. Science 168: 11591970. MECHOULAM, R.: Recent advantages in cannabinoid research. Forsch. Komplementarmed. 6 Suppl 3: 16-20, 1999. MELCK, D., RUEDA, D., GALVE, R., l, DE-PETROCELLIS, L., AND GUZMAN, M. : Involvement of the CAMP/protein kinase A pathway and of mitogen- activated protein kinase in the anti-proliferative effects of anandamide in human breast cancer cells. FEBS Lett. 463: 235-240, 1999. MENDELSON, J.H. AND IVIELLO, N.K.: Reinforcing properties of oral delta 9- tetrahydrocannabinol, smoked marijuana, and nabilone: influence of previous marijuana use. Psychopharmacology Earl 83: 351-356, 1984. MILLER, C.P., REAPE, T.J. AND FISCHER, L.J.: Inhibition of insulin production by cyproheptadine in RINm5F rat insulinoma cells. J.Biochem.Toxicol. 8: 127-134. 1993. MIURA, A., ISHIZUKA, T., ITAYA, S., ISHIZAWA, M., KANOH, Y., KIMURA, M., KAJITA, K. AND YASUDA, K.: Glucose- and phorbol ester-induced Insulin secretion in human insulinoma cells-association with protein kinase C activation. Biochem.Mol.Biol.lnt. 46: 739-745, 1998. MOHLIG, M., WOLTER, S., MAYER, P., LANG, J., OSTERHOFF, M., HORN, P.A., SCHATZ, H. AND PFEIFFER, A.: lnsulinoma cells contain an 142 Isoform of Ca2+/calmodulin-dependent protein kinase II delta associated with insulin secretion vesicles. Endocrinology 138: 2577-2584, 1997. MUGLIA, L. AND LOCKER, J.: Extrapancreatic insulin gene expression in the fetal rat. Proc.Natl.Acad.Sci.U.S.A. 81: 3635-3639, 1984. MUNRO, S., THOMAS, K. AND ABU-SHAAR, M.: Molecular Characterization of a peripheral receptor for cannabinoids . Nature 365: 61-5, 1993. NAIRN, A.C. AND PICCIOTTO, M.R.: CalciumlcalmodulIn-dependent protein kinases. Semin.Cancer Biol. 5: 295-303, 1994. NAKAMURA, M., NAGAFUCHI, S., YAMAGUSHI, K. AND TAKKI, R.: The role of thymic immunity and insulitis in the development of streptozotocin-Induced diabetes in mice. Diabetes 34: 894-900, 1984. NAKHOODA, A., LIKE, A., CHAPPEL, C., MURRAY, F. AND MARLISS, E.: The spontaneously diabetic Wistar rat. Metabolic and morphologic studies. Diabetes 26: 100-112, 1977. NELSON, K., WALSH, D., DEETER, P. AND SHEEHAN, F.: A phase II study of delta-9-tetrahydrocannabinol for appetite stimulation in cancer-associated anorexia. J.Palliat.Care 10: 14-18, 1994. NEWTON, C., KLEIN, T. AND FRIEDMAN, H.: Secondary immunity to Iegionella pneumophila and Th1 activity are suppressed by delta-9- tetrahydrocannabinol injection. Infect immun 62: 4015-4021, 1994. NEYE, H. AND VERSPOHL, E.J.: The specificity of tyrosine kinase inhibitors: their effect on insulin release (short-term effect) and insulin mRNA (long- term effect) in an insulin-secreting cell line (INS-1). Exp.Clin.Endocrinol.Diabetes 106: 292-298, 1998. NIELSEN, D., WELSH, M., CASADABAN, M. AND STEINER, 0.: Control of insulin gene expression in pancreatic beta-cells and in an insulin- producing cell line, RIN-5F cells. I. Effects of glucose and cyclic AMP on the transcription of insulin mRNA. J Biol Chem 260: 13585-13589, 1985. NIKI, I., OKAZAKI, K., SAITOH, M., NIKI, A., NIKI, H., TAMAGAWA, T., IGUCHI, A. AND HIDAKA, H.: Presence and possible involvement of Ca/calmodulin-dependent protein kinases In insulin release from the rat pancreatic beta cell. Biochem.Biophys.Res.Commun. 191: 255-261, 1993. NORLING, L.L., COLCA, J.R., KELLY, P.T., MCDANIEL, M.L. AND LANDT, M.: Activation of calcium and calmodulin dependent protein kinase II during stimulation of insulin secretion. Cell Calcium 16: 137-150, 1994. OHTA, M., NELSON, J., NELSON, D., MEGLASSON, MD. AND ERECINSKA, 143 M.: Effect of Ca++ channel blockers on energy level and stimulated insulin secretion in isolated rat islets of Langerhans. J.Pharmacol.Exp.Ther. 264: 35-40, 1993. OKAMOTO, H.: Regulation of proinsulin synthesis In pancreatic islets and a new aspect to Insulin-dependent diabetes. Mol Cell Biochem 37: 431981. OKAMOTO, H.: Molecular basis of experimental diabetes: degeneration, oncogenesis, and regeneration of pancreatic B-cells of islets of Langerhans. Bioassays 2: 151985. OSCHILEWSKI, M., SCHWAB, E., KIESEL, U., OPITZ, U., STUNKEL, K., KOLB-BACHOFEN, V. AND KOLB, H.: Administration of silica or monoclonal antibody to Thy-1 prevents low-dose streptozotocin-induced diabetes in mice. Immunol Lett 12: 289-294, 1986. PAIK, S., BLUE, M., FLEISCHER, N. AND SHIN, S.: Daibetes susceptibility of BALB/CBOM mice treated with streptozotocin. Inhibition by lethal irradition and restoration by splenic lymphocytes. Diabetes 31: 808-815, 1982. PAIK, S., MICHELIS, M., KIM, Y. AND SHIN, S.: Induction of insulin- dependentdiabetes by streptozotocin: inhibition by estrogens and potentiation by androgens. Diabetes 31: 724-729, 1982. PAN, X., IKEDA, S.R. AND LEWIS, D.L.: Rat brain cannabinoid receptor modulates N-type Ca2+ channels in a neuronal expression system. Mol.Pharmacol. 49: 707-714, 1996. PERMUTT, M.A., KAKITA, K., MALINAS, P., KARL, |., BONNER, W.S., WEIR, G. AND GIDDINGS, S.J.: An in vivo analysis of pancreatic protein and insulin biosynthesis In a rat model for non-insulin-dependent diabetes. J.CIin.Invest. 73: 1344-1350, 1984. PERMUTT, M.A. AND KIPNIS, D.M.: lnsUlIn biosynthesis. I. On the mechanism of glucose stimulation. J.Biol.Chem. 247: 1194-1199, 1972. PERMUTT.M.A: Biosynthesis of insulin. In The islets of Langerhans: Biochemistry, Physiology, and Pathology, ed. by Cooperstein.S.J and Watkins.D., pp. 75-95, Academic Press, New York, NY, 1981. PRENTKI, M., BRUN, T. AND ROCHE, E.: Nutriments, insulin secretion and gene expression in the pancreatic beta cell. Joum.Annu.Diabetol.Hotel.Dieu. 1-11: -11,1994. PERTWEE,R.G.: Pharmacology of cannabinoid CB1 and CB2 receptors. Pharmacol.Ther. 74: 129-180, 1997. PERTWEE, R.G.: Pharmacological, physiological and clinical implications of the 144 discovery of cannabinoid receptors. Biochem.Soc.Trans. 26: 267-272, 1998. PERTWEE, R.: The central neurophamacology of psychotropic cannabinoids. Pharmacol Ther 36: 189-261, 1988. PHELPS, OF. AND SAWYER, C.H.: Electrochemically stimulated release of luteinizing hormone and ovulation after surgical interruption of lateral hypothalamic connections in the rat. Brain Res. 131: 335-344, 1977. PORKSEN, N.K., MUNN, S.R., STEERS, J.L., SCHMITZ, O., VELDHUIS, J.D. AND BUTLER, P.C.: Mechanisms of sulfonylurea's stimulation of insulin secretion in vivo: selective amplification of insulin secretory burst mass. Diabetes 45: 1792-1797, 1996. PRENTKI, M., TORNHEIM, K. AND CORKEY, B.E.: Signal transduction mechanisms in nutrient-induced insulin secretion. Diabetologia 40 Suppl 2: $32-$41 1997. PROSS, S., NEWTON, C., KLEIN, T. AND FRIEDMAN, H.: Age-associated differences in cannabinoid-induced suppression of murine spleen,lymph node,and thymus cell blstogenic responses. lmmunophannacology 14: 1591987. ‘ PROSSER, P. AND KARAM, J.: Diabetes mellitus following rodenticide Ingestion In man. JAMA 239: 1148-1150, 1978. PUKEL, C., BAQUERIZO, H. AND RABINOVITCH, A.: Destruction of rat Islet cell monolayers by cytokines. Synergistic interactions of interferon-gamma, tumor necrosis factor, lymphotoxin, and interleukin 1. Diabetes 37: 133- 136, 1988. RABINOVITCH, A.: Immunoregulatory and cytokine Imbalances in the pathogenesis of IDDM. Diabetes 43: 613-621, 1994. RABINOVITCH, A., SUAREZ-PINZON, W., EL-SHEIKH, A., SORENSEN, O. AND POWER, R.: Cytokine gene expression In pancreatic islet-infiltrating leukocytes of BB rats: expression of Th1 cytokines correlates with beta- cell destructive insulitis and IDDM. Diabetes 45: 749-754, 1996. RAFIQ, I., DA, 8., X, HOOPER, S. AND RU'I'I'ER, G.A.: Glucose-stimulated preproinsulin gene expression and nuclear trans-location of pancreatic duodenum homeobox-1 require activation of phosphatidylinositol 3-kinase but not p38 MAPK/SAPK2. J.Biol.Chem. 275: 15977-15984, 2000. RAKIETEN, N., RAKIETEN, M. AND NADKARINI, M.: Studies on diabetogenic actions of streptozotocin (NSC 37917). Cancer Chemother Rep 29: 91-98, 145 1963. RAZDAN, R.K.: Structure-activity relationships in cannabinoids. Pharmacol.Rev. 38: 75-149, 1986. REDDY, S. AND SANDLER, S.: Age-dependent sensitivity to streptozotocin of pancreatic islets isolated from female NOD mice. Autoimmunity. 22: 121- 126, 1995. REIBAUD, M., OBINU, M.C., LEDENT, C., PARMENTIER, M., BOHME, G.A. AND IMPERATO, A.: Enhancement of memory in cannabinoid CB1 receptor knock-out mice. Eur.J Pharmacol. 379: R1-R21999. RINALDI, C.M., BARTH, F., HEAULME, M., SHIRE, D., CALANDRA, B., CONGY, C., MARTINEZ, S., MARUANI, J., NELIAT, G., CAPUT, D. AND ET, a.: SR141716A, a potent and selective antagonist of the brain cannabinoid receptor. FEBS Lett. 350: 240-244, 1994. RINALDI, C.M., BARTH, F., MILLAN, J., DEROCQ, J.M., CASELLAS, P., CONGY, C., OUSTRIC, D., SARRAN, M., BOUABOULA, M., CALANDRA, B., PORTIER, M., SHIRE, D., BRELIERE, J.C. AND LE- FUR, G.L.: SR 144528, the first potent and selective antagonist of the CB2 cannabinoid receptor. J Pharmacol.Exp.Ther. 284: 644-650, 1998. ROENFELDT, M., SAFAYHI, H. AND AMMON, H.P.: KCI-induced insulin secretion from RINm5F cells is mediated through Ca2+ Influx along L-type Ca2+ Channels. Naunyn Schmiedebergs Arch.Pharmacol. 346: 527-531, 1992. ROSSINI, A., LIKE, A., DULIN, W. AND CAHILL, G.J.: Pancreatic beta cell toxicity by streptozotocin anomers. Diabetes 26: 1120-1141, 1977. ROSSINI, A., WILLIAMS, R., APPEL, M. AND LIKE, A.: Complete protection from low-dose streptozotocin-induced diabetes in mice. Nature 273: 182- 184, 1978. ROSSINI, A., WILLIAMS, R., APPEL, N. AND LIKE, A.: Sex differences In the multiple-dose streptozotocin model of diabetes. Endocrinology 103: 1518- 1520, 1978. SAFAYHI, H., HAASE, H., KRAMER, U., BIHLMAYER, A., ROENFELDT, M., AMMON, H.P., FROSCHMAYR, M., CASSIDY, T.N., MORANO, l., AHLIJANIAN, M.K. AND STRIESSNIG, J.: L-type calcium Channels in insulin-secreting cells: biochemical characterization and phosphorylation in RINm5F cells. Mol.Endocrinol. 11: 619-629, 1997. SAKUMA, N., ISHIKAWA, S., OKADA, K., MIYAZAKI, J. AND SAITO, T.: Glucose induces calcium-dependent and calcium-Independent insulin 146 secretion from the pancreatic beta cell line MIN6. Eur.J.EndocrInol. 133: 227-234, 1995. SARVETNICK, N., SHIZURU, J., LIGGITI', D., MARTIN, L., MCINTYRE, B., GREGORY, A., PARSLOW, T. AND STEWART, T.: Loss of pancreatic islet tolerance induced by beta-cell expression of interferon-gamma. Nature 346: 844-871, 1990. SCANLAN, R.A.: Formation and occurrence of nitrosamines In food. Cancer Res. 43: 24353-24403, 1983. SCHARTZ, A., KESSELER, F. AND KAMINSKI, N.: Inhibition of adenylate cyclase by A9-tetrahydrocannabinol in mouse spleen cells: A potential mechanism for cannabinoid-mediated immunosuppression. Life Sci 51: 25-30, 1992. SCHARTZ, A., KOH, W. AND KAMINSKI, N.: A9-tetrahydrocannabinol selectively inhibits T-Cell dependent humoral Immune responses through direct inhibition of accessory T-Cell function. lmmunophannacology 26: 129, 1993. SCHATZ, A., LEE, M., CONDIE, R., PULASKI-JT AND KAMINSKI, N.: Cannabinoid receptors CB1 and CB2: a characterization of expression and adenylate cyclase modulation within the immune system. Toxicol Appl Pharmcol 142: 278-287, 1997. SCHEIN, P. AND BATES, R.: Plasma glucose levels in normal and adrenalectomized mice treated with streptozotocin and nicotinamide. Diabetes 17: 7601968. SELDEN, R.F., SKOSKIEWICZ, M.J., RUSSELL, P.S. AND GOODMAN, H.M.: Regulation of insulin-gene expression. Implications for gene therapy. N.EngI.J.Med. 317: 1067-1076, 1987. SERI, K., SANAI, K., KURASHIMA, K., IMAMURA, Y. AND AKITA, H.: (R)-ACX is a novel sufonylurea compound with potent, quick and short-lasting hypoglycemic activity. Eur.J.Pharmacol. 389: 253-256, 2000. SESTIER, C., ODENT-POGU, S., BONNEVILLE, M., MAUREL, C., LANG, F. AND SAI, P.: Cyclosporin enhances diabetes induced by low-dose streptozotocin treatment In mice. Immunol Lett 10: 57-60, 1985. SHEN, M. AND THAYER, SA: The cannabinoid agonist Win55,212-2 Inhibits calcium Channels by receptor-mediated and direct pathways in cultured rat hippocampal neurons. Brain Res. 783: 77-84, 1998. SHI, Z.Q., RASTOGI, K.S., LEKAS, M., EFENDIC, S., DRUCKER, D.J. AND 147 VRANIC, M.: Glucagon response to hypoglycemia is improved by Insulin- independent restoration of normoglycemia in diabetic rats. Endocrinology 137: 3193-3199, 1996. SHIVACHAR, A.C., MARTIN, B.R. AND ELLIS, E.F.: Anandamide— and delta9- tetrahydrocannabinoI-evoked arachidonic acid mobilization and blockade by SR141716A [N-(PIperidin-1-yI)-5-(4-chlorophenyI)-1-(2,4- dichlorophenyl)-4 -methyI-1H-pyrazoIe-3-carboximide hydrochloride]. Biochem.Pharmacol. 51: 669-676, 1996. SMITH, C.G., SMITH, M.T., BESCH, N.F., SMITH, R.G. AND ASCH, R.H.: Effect of delta 9-tetrahydrocannabinol (THC) on female reproductive function. Adv.Biosci. 22-23: 449-467, 1978. SOBIESZEK, A.: Regulation of myosin light Chain kinase: kinetic mechanism, autophosphorylation, and cooperative activation by Ca2+ and calmodulin. Can.J.PhysIol.Pharmacol. 72: 1368-1376, 1994. STEINER, D.F.: Evidence for a precursor in the biosynthesis of insulin. Trans.N.Y.Acad.SCI. 30: 60-68, 1967. STEINER, D.F., CHAN, S.J., WELSH, J.M. AND KWOK, 8.0.: Structure and evolution of the insulin gene. Annu.Rev.Genet. 19: 463—484, 1985. STEINER, D.F. AND TAGER, H.S.: Biosynthesis of insulin and glucagon. In Endocrinology, ed. by DeGroot, pp. 921-934, Grunne and Sytatton, Inc., New York, NY, 1979. STERNESJO, J., BENDTZEN, K. AND SANDLER, 8.: Effects of prolonged exposure in vitro to interferon-gamma and tumour necrosis factor-alpha on nitric oxide and insulin production of rat pancreatic islets. Autoimmunity. 20: 185-190, 1995. SUGIURA, T., KODAKA, T., KONDO, S., NAKANE, S., KONDO, H., WAKU, K., ISHIMA, Y., WATANABE, K. AND YAMAMOTO, I.: Is the cannabinoid CB1 receptor a 2-arachidonoylglycerol receptor? Structural requirements for triggering a Ca2+ transient in NG108-15 cells. J.Biochem.Tokyo. 122: 890-895, 1997. TAKASAWA, S., AKIYAMA, T., NATA, K., KUROKI, M., TOHGO, A., NOGUCHI. N., KOBAYASHI, S., KATO, I., KATADA, T. AND OKAMOTO, H.: Cyclic ADP-ribose and inositol 1,4,5-trisphosphate as alternate second messengers for intracellular Ca2+ mobilization in normal and diabetic beta-cells. J.Biol.Chem. 273: 2497-2500, 1998. TAMURA, K., FUJIMURA, T., TSUTSUMI, T., NAKAMURA, K., OGAWA, T., ATUMARU, C., HIRANO, Y., OHARA, K., OHTSUKA, K., SHIMOMURA, K. AND ET, a.: Inhibition of insulin production by FK 506 is caused at the 148 transcriptional level in pancreatic beta cell when FK BP-12 content is relatively high. Transplant.Proc. 27: 357-361, 1995. TABUCHI, H., YAMAMOTO, H., MATSUMOTO, K., EBIHARA, K., TAKEUCHI, Y., FUKUNAGA, K., HIRAOKA, H., SASAKI, Y., SHICHIRI, M. AND MIYAMOTO, E.: Regulation of insulin secretion by overexpression of Ca2+/calmodulIn-dependent protein kinase II In Insulinoma MIN6 cells. Endocrinology 141: 2350-2360, 2000. TANG, S.H. AND SHARP, G.W.: Atypical protein kinase C Isozyme zeta mediates carbachol-stimulated insulin secretion in RINm5F cells. Diabetes 47: 905-912, 1998. TANG, S.H., YANEY, G.C. AND SHARP, G.W.: Unusual carbachol responses in RINm5F cells: evidence for a "distal" site of action in stimulus-secretion coupling. Mol.Pharmacol. 47: 863-870, 1995. TAYLOR, H.G.: Analysis of the medical use of marijuana and its societal Implications [see comments]. J.Am.Pharm.Assoc.Wash. 38 : 220-227, 1998. THOMAS, B.F., COMPTON, D.R. AND MARTIN, B.R.: Characterization of the lipophilicity of natural and synthetic analogs of delta 9- tetrahydrocannabinol and its relationship to pharmacological potency. J.Pharmacol.Exp.Ther. 255: 624-630, 1990. TJALVE, H.: Streptozotocin: distribution, metabolism, and mechanisms of action. Ups J Med Sci [suppl]39: 145-157, 1983. TREMBLEAU, S., GERMANN, T., GATELY, M. AND ADORINI, L.: The role of IL- 12 In the induction of organ-specific autoimmune diseases. Immunol Today 16: 383-386, 1995. TWITCHELL, W., BROWN, S. AND MACKIE, K.: Cannabinoids inhibit N- and P/Q-type calcium Channels in cultured rat hippocampal neurons. J Neurophysiol. 78: 43—50, 1997. UNGERLEIDER, J. AND ANDRYSIAK, T.: Therapeutic issues of marijuana and THC (tetrahydrocannabinol). Int J Addict 20: 691-699, 1985. VELEMINSKY, J., BURR, I.M. AND STAUFFACHER, W.: Comparative study of ear1y metabolic events resulting from the administration of the two diabetogenic agents alloxan and streptozotocin. Eur.J.CIin.lnvest. 1: 104- 108, 1970. VOGEL, Z., BARG, J., LEW, R., SAYA, D., HELDMAN, E. AND MECHOULAM, R.: Anandamide, a brain endogenous compound, interacts specifically with cannabinoid receptors and Inhibits adenylate cyclase. J Neurochem. 149 61: 352-355, 1993. VOTH, E. AND SCHWARTZ, R.: Medicinal applications of delta-9- tetrahydrocannabinol and marijuana. Ann lntem Med 126: 791-798, 1997. WALSH, M.P., DABROWSKA, R., HINKINS, S. AND HARTSHORNE, D.J.: Calcium-independent myosin light chain kinase of smooth muscle. Preparation by limited chymotryptic digestion of the calcium ion dependent enzyme, purification, and Characterization. Biochemistry 21: 1919-1925, 1982. WEBER, C., PERNIS, B. AND REEMTSMA, K.: Major histocompatibility complex,genetic background and streptozotocin diabetes. Surg Forum 34: 365-367, 1983. WEBER, C., PERNIS, B., TING, W., ROSENKRANTZ, K. AND REEMTSMA, K.: Murine streptozotocin diabetes: influences of the major histocompatibility complex, genetic background and blood transfusion. Diabetologia 27: 160-162, 1984. WELCH, S.P., HUFFMAN, J.W. AND LOWE, J.: Differential blockade of the antinociceptive effects of centrally administered cannabinoids by SR141716A. J Pharmacol.Exp.Ther. 286: 1301-1308, 1998. WELSH, M., BRUNSTEDT, J. AND HELLERSTROM, C.: Effects of D-glucose, L- leucine, and 2-ketoisocaproate on insulin mRNA levels in mouse pancreatic islets. Diabetes 35: 228-231, 1986. WELSH, M., SCHERBERG, N., GILMORE, R. AND STEINER, D.F.: Translational control of Insulin biosynthesis. Evidence for regulation of elongation, initiation and signal-recognition-particIe-mediated translational arrest by glucose. Biochem.J. 235: 459-467, 1986. WELSH, M., WEBER, T., WRANGE, O., NIELSEN, D., MATTHIEU, M. AND STEINER, D.: Regulation of insulin gene expression by dexamethasone, Ca2+ and a phorbol ester. Biomed Biochim Acta 47: 299-303, 1988. WENGER, T., TOTH, B.E., JUANEDA, C., LEONARDELLI, J. AND TRAMU, G.: The effects of cannabinoids on the regulation of reproduction. Life Sci. 65: 695-701, 1999. WENHAM, R.M., LANDT, M. AND EASOM, R.A.: Glucose activates the multifunctional CaZ+/calmodulin-dependent protein kinase II in isolated rat pancreatic islets. J.Biol.Chem. 269: 4947-4952, 1994. WIGGANS, R., WOOLEY, P., MACDONALD, J., SMYTHE, T., UENO, W. AND SCHEIN, P.: Phase II trial of streptozotocin, mltomycin-c, and 5- fluorouracil (SMF) in treatment of advanced pancreatic cancer. Cancer 41: 150 387-391, 1959. WILSON, G., HARTIG, P., PATTON, N. AND LEDOUX, S.: Mechanisms of nitrourea-induced B cell damage: activation of poly (ADP-ribose) synthetase and cellular distribution. Diabetes 37: 213—216, 1988. WIRGUIN, I., MECHOULAM, R., BREUER, A., SCHEZEN, E., WEIDENFELD, J. AND BRENNER, T.: Syppression of experimental autoimmune encephalomylitis by cannabinoids. lmmunophannacology 28: 209-214, 1994. WOLF, J., LILLY, F. AND SHIN, S.: The influence of genetic background on the susceptibility on inbred mice to streptozotocin-induced diabetes. Diabetes 33: 567-571, 1984. WOLLHEIM, C.B. AND POZZAN, T.: Correlation between cytosolic free Ca2+ and insulin release in an insulin-secreting cell line. J.Biol.Chem. 259: 2262-2267, 1984. YAMAMOTO, h., UCHIGATA, Y. AND OKAMOTO, H.: Streptozotocin and alloxan induce DNA strand breaks and poly(ADP-ribose) synthetase in pancreatic Islets. Nature 294: 284-286, 1981. YONEMURA, Y., TASKASHIMA, T., MIWA, K., MIYAZAKI, I., YAMAMOTO, h. AND OKAMATO, H.: Amelioration of diabetes mellitus In partially depancreatized rats by poly(ADP-ribose) synthetase inhibitors: evidence of islet B-cell regeneration. Diabetes 33: 401-404, 1984. YOON, J.: Role of viruses and environmental factors in induction of diabetes. In Human diabetes: Genetic, environmental and Autoimmune Etiology. ed. by B.-S. Hansen-B, pp. 95-123, Springer-Venag, New York, 1990. ZAWALICH, W.S., BONNET, E.M., ZAWALICH, K.C. AND YANEY, G.C.: Chronic exposure to TPA depletes PKC alpha and augments Ca- dependent insulin secretion from cultured rat islets. Am.J.Physiol. 274: C1 388-C13961 998. ZHENG, Z., SPECTER, S. AND FRIEDMAN, H.: Inhibition by delta-9- tetrahydrocannabinol of tumor necrosis factor alpha production by mouse and human macrophages. Int J lmmunopharmacol 14: 1445-1452, 1992. 151