Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters










Publication year range
1.
Mol Metab ; 78: 101822, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37838014

ABSTRACT

OBJECTIVE: Pro-inflammatory polarization of adipose tissue macrophages (ATMs) plays a critical role in the pathogenesis of obesity-associated chronic inflammation. However, little is known about the role of lipids in the regulation of ATMs polarity and inflammation in response to metabolic stress. Deletion of α/ß-hydrolase domain-containing 6 (ABHD6), a monoacylglycerol (MAG) hydrolase, has been shown to protect against diet-induced obesity and insulin resistance. METHODS: Here we investigated the immunometabolic role of macrophage ABHD6 in response to nutrient excess using whole-body ABHD6-KO mice and human and murine macrophage cell-lines treated with KT203, a selective and potent pharmacological ABHD6 inhibitor. RESULTS: KO mice on high-fat diet showed lower susceptibility to systemic diet-induced inflammation. Moreover, in the setting of overnutrition, stromal vascular cells from gonadal fat of KO vs. control mice contained lower number of M1 macrophages and exhibited enhanced levels of metabolically activated macrophages (MMe) and M2 markers, oxygen consumption, and interleukin-6 (IL-6) release. Likewise, under in vitro nutri-stress condition, inhibition of ABHD6 in MMe-polarized macrophages attenuated the expression and release of pro-inflammatory cytokines and M1 markers and induced the upregulation of lipid metabolism genes. ABHD6-inhibited MMe macrophages showed elevated levels of peroxisome proliferator-activated receptors (PPARs) and 2-MAG species. Notably, among different MAG species, only 2-MAG treatment led to increased levels of PPAR target genes in MMe macrophages. CONCLUSIONS: Collectively, our findings identify ABHD6 as a key component of pro-inflammatory macrophage activation in response to excess nutrition and implicate an endogenous macrophage lipolysis/ABHD6/2-MAG/PPARs cascade, as a lipid signaling and immunometabolic pathway, which favors the anti-inflammatory polarization of ATMs in obesity.


Subject(s)
Monoglycerides , Peroxisome Proliferator-Activated Receptors , Humans , Animals , Mice , Peroxisome Proliferator-Activated Receptors/metabolism , Monoglycerides/metabolism , Mice, Obese , Hydrolases/genetics , Hydrolases/metabolism , Adipose Tissue/metabolism , Macrophages/metabolism , Obesity/metabolism , Inflammation/metabolism , Anti-Inflammatory Agents , Diet, High-Fat/adverse effects , Monoacylglycerol Lipases/genetics , Monoacylglycerol Lipases/metabolism
2.
Diabetologia ; 56(6): 1394-402, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23508306

ABSTRACT

AIMS/HYPOTHESIS: Lysophosphatidic acid (LPA) is a lipid mediator produced by adipocytes that acts via specific G-protein-coupled receptors; its synthesis is modulated in obesity. We previously reported that reducing adipocyte LPA production in high-fat diet (HFD)-fed obese mice is associated with improved glucose tolerance, suggesting a negative impact of LPA on glucose homeostasis. Here, our aim was to test this hypothesis. METHODS: First, glucose tolerance and plasma insulin were assessed after acute (30 min) injection of LPA (50 mg/kg) or of the LPA1/LPA3 receptor antagonist Ki16425 (5 mg kg(-1) day(-1), i.p.) in non-obese mice fed a normal diet (ND) and in obese/prediabetic (defined as glucose-intolerant) HFD mice. Glucose and insulin tolerance, pancreas morphology, glycogen storage, glucose oxidation and glucose transport were then studied after chronic treatment (3 weeks) of HFD mice with Ki16425. RESULTS: In ND and HFD mice, LPA acutely impaired glucose tolerance by inhibiting glucose-induced insulin secretion. These effects were blocked by pre-injection of Ki16425 (5 mg/kg, i.p.). Inhibition of glucose-induced insulin secretion by LPA also occurred in isolated mouse islets. Plasma LPA was higher in HFD mice than in ND mice and Ki16425 transiently improved glucose tolerance. The beneficial effect of Ki16425 became permanent after chronic treatment and was associated with increased pancreatic islet mass and higher fasting insulinaemia. Chronic treatment with Ki16425 also improved insulin tolerance and increased liver glycogen storage and basal glucose use in skeletal muscle. CONCLUSIONS/INTERPRETATION: Exogenous and endogenous LPA exerts a deleterious effect on glucose disposal through a reduction of plasma insulin; pharmacological blockade of LPA receptors improves glucose homeostasis in obese/prediabetic mice.


Subject(s)
Diet, High-Fat , Gene Expression Regulation , Glucose/metabolism , Insulin/metabolism , Lysophospholipids/metabolism , Adipocytes/cytology , Animals , Body Weight , Glycogen/metabolism , Homeostasis , Insulin Secretion , Isoxazoles/pharmacology , Lipids/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle, Skeletal/metabolism , Oxygen/metabolism , Propionates/pharmacology , Time Factors
3.
Diabetologia ; 55(10): 2682-2692, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22820510

ABSTRACT

AIMS/HYPOTHESIS: Activation of the G protein-coupled receptor (GPR)40 by long-chain fatty acids potentiates glucose-stimulated insulin secretion (GSIS) from pancreatic beta cells, and GPR40 agonists are in clinical development for type 2 diabetes therapy. GPR40 couples to the G protein subunit Gα(q/11) but the signalling cascade activated downstream is unknown. This study aimed to determine the mechanisms of GPR40-dependent potentiation of GSIS by fatty acids. METHODS: Insulin secretion in response to glucose, oleate or diacylglycerol (DAG) was assessed in dynamic perifusions and static incubations in islets from wild-type (WT) and Gpr40 (-/-) mice. Depolymerisation of filamentous actin (F-actin) was visualised by phalloidin staining and epifluorescence. Pharmacological and molecular approaches were used to ascertain the roles of protein kinase D (PKD) and protein kinase C delta in GPR40-mediated potentiation of GSIS. RESULTS: Oleate potentiates the second phase of GSIS, and this effect is largely dependent upon GPR40. Accordingly, oleate induces rapid F-actin remodelling in WT but not in Gpr40 (-/-) islets. Exogenous DAG potentiates GSIS in both WT and Gpr40 (-/-) islets. Oleate induces PKD phosphorylation at residues Ser-744/748 and Ser-916 in WT but not Gpr40 (-/-) islets. Importantly, oleate-induced F-actin depolymerisation and potentiation of GSIS are lost upon pharmacological inhibition of PKD1 or deletion of Prkd1. CONCLUSIONS/INTERPRETATION: We conclude that the signalling cascade downstream of GPR40 activation by fatty acids involves activation of PKD1, F-actin depolymerisation and potentiation of second-phase insulin secretion. These results provide important information on the mechanisms of action of GPR40, a novel drug target for type 2 diabetes.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Protein Kinase C/physiology , Receptors, G-Protein-Coupled/physiology , Actins/metabolism , Animals , Cells, Cultured , Diglycerides/pharmacology , Glucose/pharmacology , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Mice , Mice, Knockout , Models, Animal , Oleic Acid/pharmacology , Protein Kinase C-delta/deficiency , Protein Kinase C-delta/genetics , Protein Kinase C-delta/physiology , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Signal Transduction/physiology
4.
Diabetologia ; 53(11): 2369-79, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20628728

ABSTRACT

AIMS/HYPOTHESIS: Prolonged exposure of pancreatic beta cells to excessive levels of glucose and fatty acids, referred to as glucolipotoxicity, is postulated to contribute to impaired glucose homeostasis in patients with type 2 diabetes. However, the relative contribution of defective beta cell function vs diminished beta cell mass under glucolipotoxic conditions in vivo remains a subject of debate. We therefore sought to determine whether glucolipotoxicity in rats is due to impaired beta cell function and/or reduced beta cell mass, and whether older animals are more susceptible to glucolipotoxic condition. METHODS: Wistar rats (2 and 6 months old) received a 72 h infusion of glucose + intravenous fat emulsion or saline control. In vivo insulin secretion and sensitivity were assessed by hyperglycaemic clamps. Ex vivo insulin secretion, insulin biosynthesis and gene expression were measured in isolated islets. Beta cell mass and proliferation were examined by immunohistochemistry. RESULTS: A 72 h infusion of glucose + intravenous fat emulsion in 2-month-old Wistar rats did not affect insulin sensitivity, insulin secretion or beta cell mass. In 6-month-old rats by contrast it led to insulin resistance and reduced insulin secretion in vivo, despite an increase in beta cell mass and proliferation. This was associated with: (1) diminished glucose-stimulated second-phase insulin secretion and proinsulin biosynthesis; (2) lower insulin content; and (3) reduced expression of beta cell genes in isolated islets. CONCLUSIONS/INTERPRETATION: In this in vivo model, glucolipotoxicity is characterised by an age-dependent impairment of glucose-regulated beta cell function despite a marked increase in beta cell mass.


Subject(s)
Fatty Acids/toxicity , Glucose/toxicity , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Immunohistochemistry , In Vitro Techniques , Insulin/metabolism , Insulin-Secreting Cells/pathology , Male , Proinsulin/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
5.
Diabetologia ; 52(6): 1122-32, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19294363

ABSTRACT

AIMS/HYPOTHESIS: The Zucker fatty (ZF) rat subjected to 60% pancreatectomy (Px) develops moderate diabetes by 3 weeks. We determined whether a progressive fall in beta cell mass and/or beta cell dysfunction contribute to beta cell failure in this type 2 diabetes model. METHODS: Partial (60%) or sham Px was performed in ZF and Zucker lean (ZL) rats. At 3 weeks post-surgery, beta cell mass and proliferation, proinsulin biosynthesis, pancreatic insulin content, insulin secretion, and islet glucose and lipid metabolism were measured. RESULTS: ZL-Px rats maintained normal glycaemia and glucose-stimulated insulin secretion (GSIS) despite incomplete recovery of beta cell mass possibly due to compensatory enhanced islet glucose metabolism and lipolysis. ZF-Px rats developed moderate hyperglycaemia (14 mmol/l), hypertriacylglycerolaemia and relative hypoinsulinaemia. Despite beta cell mass recovery and normal arginine-induced insulin secretion, GSIS and pancreatic insulin content were profoundly lowered in ZF-Px rats. Proinsulin biosynthesis was not reduced. Compensatory increases in islet glucose metabolism above those observed in ZF-Sham rats were not seen in ZF-Px rats. Triacylglycerol content was not increased in ZF-Px islets, possibly due to lipodetoxification by enhanced lipolysis and fatty acid oxidation. Fatty acid accumulation into monoacylglycerol and diacylglycerol was increased in ZF-Px islets together with a 4.5-fold elevation in stearoyl-CoA desaturase mRNA expression. CONCLUSIONS/INTERPRETATION: Falling beta cell mass, reduced proinsulin biosynthesis and islet steatosis are not implicated in early beta cell failure and glucolipotoxicity in ZF-Px rats. Rather, severe beta cell dysfunction with a specific reduction in GSIS and marked depletion of beta cell insulin stores with altered lipid partitioning underlie beta cell failure in this animal model of type 2 diabetes.


Subject(s)
Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Obesity/metabolism , Obesity/pathology , Animals , Body Weight , Cell Proliferation , Cells, Cultured , Fatty Acids, Nonesterified/metabolism , Hyperlipidemias/physiopathology , Immunohistochemistry , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Lipid Metabolism/physiology , Male , Obesity/physiopathology , Pancreatectomy , Proinsulin/metabolism , Rats , Rats, Zucker
6.
Diabetologia ; 52(4): 723-32, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19169664

ABSTRACT

AIMS/HYPOTHESIS: The molecular mechanisms by which thiazolidinediones improve insulin sensitivity in type 2 diabetes are not fully understood. We hypothesised that pioglitazone would activate the adenosine 5'-monophosphate-activated protein kinase (AMPK) pathway and increase the expression of genes involved in adiponectin signalling, NEFA oxidation and mitochondrial function in human skeletal muscle. METHODS: A randomised, double-blind, parallel study was performed in 26 drug-naive type 2 diabetes patients treated with: (1) pioglitazone (n = 14) or (2) aggressive nutritional therapy (n = 12) to reduce HbA(1c) to levels observed in the pioglitazone-treated group. Participants were assigned randomly to treatment using a table of random numbers. Before and after 6 months, patients reported to the Clinical Research Center of the Texas Diabetes Institute for a vastus lateralis muscle biopsy followed by a 180 min euglycaemic-hyperinsulinaemic (80 mU m(-2) min(-1)) clamp. RESULTS: All patients in the pioglitazone (n = 14) or nutritional therapy (n = 12) group were included in the analysis. Pioglitazone significantly increased plasma adiponectin concentration by 79% and reduced fasting plasma NEFA by 35% (both p < 0.01). Following pioglitazone, insulin-stimulated glucose disposal increased by 30% (p < 0.01), and muscle AMPK and acetyl-CoA carboxylase (ACC) phosphorylation increased by 38% and 53%, respectively (p < 0.05). Pioglitazone increased mRNA levels for adiponectin receptor 1 and 2 genes (ADIPOR1, ADIPOR2), peroxisome proliferator-activated receptor gamma, coactivator 1 gene (PPARGC1) and multiple genes involved in mitochondrial function and fat oxidation. Despite a similar reduction in HbA(1c) and similar improvement in insulin sensitivity with nutritional therapy, there were no significant changes in muscle AMPK and ACC phosphorylation, or the expression of ADIPOR1, ADIPOR2, PPARGC1 and genes involved in mitochondrial function and fat oxidation. No adverse (or unexpected) effects or side effects were reported from the study. CONCLUSIONS/INTERPRETATIONS: Pioglitazone increases plasma adiponectin levels, stimulates muscle AMPK signalling and increases the expression of genes involved in adiponectin signalling, mitochondrial function and fat oxidation. These changes may represent an important cellular mechanism by which thiazolidinediones improve skeletal muscle insulin sensitivity. TRIAL REGISTRATION: NCT 00816218 FUNDING: This trial was funded by National Institutes of Health Grant DK24092, VA Merit Award, GCRC Grant RR01346, Executive Research Committee Research Award from the University of Texas Health Science Center at San Antonio, American Diabetes Association Junior Faculty Award, American Heart Association National Scientist Development Grant, Takeda Pharmaceuticals North America Grant and Canadian Institute of Health Research Grant.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adiponectin/blood , Diabetes Mellitus, Type 2/drug therapy , Gene Expression Regulation/drug effects , Hypoglycemic Agents/therapeutic use , Mitochondria, Muscle/metabolism , Thiazolidinediones/therapeutic use , AMP-Activated Protein Kinases/drug effects , Blood Glucose/drug effects , Blood Glucose/metabolism , DNA Primers , Diet, Diabetic , Double-Blind Method , Fatty Acids, Nonesterified/blood , Female , Glucose Clamp Technique , Humans , Hyperinsulinism , Male , Malonyl Coenzyme A/metabolism , Middle Aged , Pioglitazone , Polymerase Chain Reaction , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
Diabetologia ; 49(9): 2120-30, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16868750

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to determine the role of fatty acid signalling in islet beta cell compensation for insulin resistance in the Zucker fatty fa/fa (ZF) rat, a genetic model of severe obesity, hyperlipidaemia and insulin resistance that does not develop diabetes. MATERIALS AND METHODS: NEFA augmentation of insulin secretion and fatty acid metabolism were studied in isolated islets from ZF and Zucker lean (ZL) control rats. RESULTS: Exogenous palmitate markedly potentiated glucose-stimulated insulin secretion (GSIS) in ZF islets, allowing robust secretion at physiological glucose levels (5-8 mmol/l). Exogenous palmitate also synergised with glucagon-like peptide-1 and the cyclic AMP-raising agent forskolin to enhance GSIS in ZF islets only. In assessing islet fatty acid metabolism, we found increased glucose-responsive palmitate esterification and lipolysis processes in ZF islets, suggestive of enhanced triglyceride-fatty acid cycling. Interruption of glucose-stimulated lipolysis by the lipase inhibitor Orlistat (tetrahydrolipstatin) blunted palmitate-augmented GSIS in ZF islets. Fatty acid oxidation was also higher at intermediate glucose levels in ZF islets and steatotic triglyceride accumulation was absent. CONCLUSIONS/INTERPRETATION: The results highlight the potential importance of NEFA and glucoincretin enhancement of insulin secretion in beta cell compensation for insulin resistance. We propose that coordinated glucose-responsive fatty acid esterification and lipolysis processes, suggestive of triglyceride-fatty acid cycling, play a role in the coupling mechanisms of glucose-induced insulin secretion as well as in beta cell compensation and the hypersecretion of insulin in obesity.


Subject(s)
Insulin Resistance , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Animals , Binding Sites , Colforsin/pharmacology , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Nonesterified/pharmacology , Gene Expression Regulation/drug effects , Glucagon-Like Peptide 1/pharmacology , Glucose/pharmacology , In Vitro Techniques , Insulin Secretion , Insulin-Secreting Cells/drug effects , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Lactones/metabolism , Lactones/pharmacology , Lipase/metabolism , Lipid Metabolism/drug effects , Lipolysis/drug effects , Models, Biological , Orlistat , Oxidation-Reduction/drug effects , Rats , Rats, Zucker , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
8.
Diabetologia ; 47(5): 806-15, 2004 May.
Article in English | MEDLINE | ID: mdl-15095038

ABSTRACT

AIMS/HYPOTHESIS: We have provided evidence that glucagon-like peptide-1, a potential therapeutic agent in the treatment of diabetes, activates phosphatidylinositol-3 kinase/protein kinase B signalling in the pancreatic beta cell. Since this pathway promotes cell survival in a variety of systems, we tested whether glucagon-like peptide-1 protects beta cells against cell death induced by elevated glucose and/or non-esterified fatty acids. METHODS: Human islets and INS832/13 cells were cultured at glucose concentrations of 5 or 25 mmol/l in the presence or absence of palmitate. Apoptosis was evaluated by monitoring DNA fragmentation and chromatin condensation. Wild-type and protein kinase B mutants were overexpressed in INS832/13 cells using adenoviruses. Nuclear factor-kappa B DNA binding was assayed by electrophoretic mobility shift assay. RESULTS: In human pancreatic beta cells and INS832/13 cells, glucagon-like peptide-1 prevented beta cell apoptosis induced by elevated concentrations of (i) glucose (glucotoxicity), (ii) palmitate (lipotoxicity) and (iii) both glucose and palmitate (glucolipotoxicity). Overexpression of a dominant-negative protein kinase B suppressed the anti-apoptotic action of glucagon-like peptide-1 in INS832/13 cells, whereas a constitutively active protein kinase B prevented beta cell apoptosis induced by elevated glucose and palmitate. Glucagon-like peptide-1 enhanced nuclear factor-kappa B DNA binding activity and stimulated the expression of inhibitor of apoptosis protein-2 and Bcl-2, two anti-apoptotic genes under the control of nuclear factor-kappa B. Inhibition of nuclear factor-kappa B by BAY 11-7082 abolished the prevention of glucolipotoxicity by glucagon-like peptide-1. CONCLUSIONS/INTERPRETATION: The results demonstrate a potent protective effect of glucagon-like peptide-1 on beta cell gluco-, lipo- and glucolipotoxicity. This effect is mediated via protein kinase B activation and possibly its downstream target nuclear factor-kappa B.


Subject(s)
Apoptosis/drug effects , Glucose/toxicity , Islets of Langerhans/cytology , Palmitic Acid/toxicity , Peptide Fragments/pharmacology , Cell Line , Cells, Cultured , Glucagon , Glucagon-Like Peptide 1 , Glucagon-Like Peptides , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitriles/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Sulfones/pharmacology
9.
Acta Physiol Scand ; 178(4): 435-42, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12864749

ABSTRACT

UNLABELLED: An increasing body of evidence has revealed that activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK)-activated protein kinase increases fatty acid oxidation by lowering the concentration of malonyl coenzyme A (CoA), an inhibitor of carnitine palmitoyl transferase 1. Studies carried out primarily in skeletal muscle suggest that AMPK modulates the concentration of malonyl CoA by concurrently phosphorylating and inhibiting acetyl CoA carboxylase (ACC), the rate limiting enzyme in malonyl CoA synthesis, and phosphorylating and activating malonyl CoA decarboxylase (MCD), an enzyme involved in its degradation. We have recently observed that AMPK and MCD activities are increased and ACC activity diminished in skeletal muscle, liver and, surprisingly, in adipose tissue 30 min following exercise (treadmill run) in normal rats. In liver and adipose tissue these changes were associated with a decrease in the activity of glycerol-3-phosphate acyltransferase (GPAT), which catalyses the first committed reaction in glycerolipid synthesis and, which like ACC, is phosphorylated and inhibited by AMPK. Similar changes in ACC, MCD and GPAT were observed following the administration of 5-aminoimidazole 4-carboxamide-riboside (AICAR), further indicating that the exercise-induced alterations in these enzymes were AMPK-mediated. CONCLUSIONS: (1) AMPK plays a major role in regulating lipid metabolism in multiple tissues following exercise. (2) The net effect of its activation is to increase fatty acid oxidation and diminish glycerolipid synthesis. (3) The relevance of these findings to the regulation of muscle glycogen repletion in the post-exercise state and to the demonstrated ability of AMPK activation to decrease adiposity and increase insulin sensitivity in rodents remains to be determined.


Subject(s)
Adipose Tissue/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Cyclic AMP-Dependent Protein Kinases/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal/physiology , Acetyl-CoA Carboxylase/metabolism , Adipose Tissue/drug effects , Aminoimidazole Carboxamide/pharmacology , Animals , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Hypoglycemic Agents/pharmacology , Liver/drug effects , Malonyl Coenzyme A/metabolism , Muscle, Skeletal/drug effects , Rats , Ribonucleotides/pharmacology
10.
Atherosclerosis ; 161(2): 345-52, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11888517

ABSTRACT

Chronic intraperitoneal or subcutaneous insulin administration increases triglyceride secretion rate (TGSR) in normal rats. We wished to determine the effect of this treatment on TGSR and the hepatic lipogenic enzymes acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS) in diabetic rats. Streptozotocin-diabetic rats, untreated (D), diabetic rats treated with insulin (3 U/day for 21 days) intraperitoneally (IP) or subcutaneously (SC) and non-diabetic rats (N) were studied. TGSR was determined using Triton WR-1339. Fasting glucose and triglyceride levels, high in D, were normalized by insulin treatment regardless of route. Peripheral insulin levels were lowest in D and highest in SC, portal insulin levels were lowest in D and highest in IP. Non-esterified fatty acid levels were not elevated in D, presumably due to adipose tissue depletion. TGSR was reduced in D (P<0.05) and was normalized following insulin administration, regardless of route. ACC activity was normal, but FAS was decreased in D (P<0.05). ACC and FAS were normal in both IP and SC. Thus, in streptozotocin-diabetic rats, chronic intraperitoneal or subcutaneous insulin treatment increases TGSR and FAS activity from their low levels in insulin-deficient rats to levels equal to but not higher than those in normal rats.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Fatty Acid Synthases/metabolism , Insulin/administration & dosage , Triglycerides/metabolism , Analysis of Variance , Animals , Disease Models, Animal , Glucose/metabolism , Injections, Intraperitoneal , Injections, Subcutaneous , Liver/enzymology , Male , Probability , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity , Streptozocin
11.
Metabolism ; 51(1): 110-4, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11782881

ABSTRACT

This study intended to test the hypothesis that intracellular lipolysis in the pancreatic beta cells is implicated in the regulation of insulin secretion stimulated by nutrient secretagogues or cyclic adenosine monophosphate (cAMP) agonists. Indeed, although lipid signaling molecules were repeatedly reported to influence beta-cell function, the contribution of intracellular triglycerides to the generation of these molecules has remained elusive. Thus, we have studied insulin secretion of isolated rat pancreatic islets in response to various secretagogues in the presence or absence of 3,5-dimethylpyrazole (DMP), a water-soluble and highly effective antilipolytic agent, as previously shown in vivo. In vitro exposure of islets to DMP resulted in an inhibition (by approximately 50%) of the insulin release stimulated not only by high glucose, but also by another nutrient secretagogue, 2-ketoisocaproate, as well as the cAMP agonists 3-isobutyl-1-methylxanthine and glucagon. The inhibitory effect of DMP, which was not due to alteration of islet glucose oxidation, could be reversed upon addition of sn-1,2-dioctanoylglycerol, a synthetic diglyceride, which activates protein kinase C. The results provide direct pharmacologic evidence supporting the concept that endogenous beta-cell lipolysis plays an important role in the generation of lipid signaling molecules involved in the control of insulin secretion in response to both fuel stimuli and cAMP agonists.


Subject(s)
Cyclic AMP/agonists , Insulin/metabolism , Islets of Langerhans/metabolism , Lipolysis/drug effects , Nutritional Physiological Phenomena , Pyrazoles/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Diglycerides/pharmacology , Glucagon/pharmacology , Glucose/pharmacology , In Vitro Techniques , Insulin Secretion , Islets of Langerhans/drug effects , Keto Acids/pharmacology , Male , Rats , Rats, Sprague-Dawley
12.
Diabetes ; 50(10): 2237-43, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11574404

ABSTRACT

Glucagon-like peptide-1 (GLP-1), an insulinotropic and glucoincretin hormone, is a potentially important therapeutic agent in the treatment of diabetes. We previously provided evidence that GLP-1 induces pancreatic beta-cell growth nonadditively with glucose in a phosphatidylinositol-3 kinase (PI-3K)-dependent manner. In the present study, we investigated the downstream effectors of PI-3K to determine the precise signal transduction pathways that mediate the action of GLP-1 on beta-cell proliferation. GLP-1 increased extracellular signal-related kinase 1/2, p38 mitogen-activated protein kinase (MAPK), and protein kinase B activities nonadditively with glucose in pancreatic beta(INS 832/13) cells. GLP-1 also caused nuclear translocation of the atypical protein kinase C (aPKC) zeta isoform in INS as well as in dissociated normal rat beta-cells as shown by immunolocalization and Western immunoblotting analysis. Tritiated thymidine incorporation measurements showed that the p38 MAPK inhibitor SB203580 suppressed GLP-1-induced beta-cell proliferation. Further investigation was performed using isoform-specific pseudosubstrates of classical (alpha, beta, and gamma) or zeta aPKC isoforms. The PKCzeta pseudosubstrate suppressed the proliferative action of GLP-1, whereas the inhibitor of classical PKC isoforms had no effect. Overexpression of a kinase-dead PKCzeta acting as a dominant negative protein suppressed GLP-1-induced proliferation. In addition, ectopic expression of a constitutively active PKCzeta mutant stimulated tritiated thymidine incorporation to the same extent as GLP-1, and the glucoincretin had no growth-promoting action under this condition. The data indicate that GLP-1-induced activation of PKCzeta is implicated in the beta-cell proliferative signal of the insulinotropic hormone. The results are consistent with a model in which GLP-1-induced PI-3K activation results in PKCzeta translocation to the nucleus, which may play a role in the pleiotropic effects (DNA synthesis, metabolic enzymes, and insulin gene expression) of the glucoincretin.


Subject(s)
Glucagon/pharmacology , Islets of Langerhans/cytology , Peptide Fragments/pharmacology , Protein Kinase C/metabolism , Protein Precursors/pharmacology , Animals , Biological Transport/drug effects , Cell Division/drug effects , Cell Division/physiology , Cell Line , Cell Nucleus/metabolism , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Glucagon-Like Peptide 1 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mutation/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Rats , Rats, Wistar , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases
13.
Diabetes ; 50(9): 1970-5, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11522661

ABSTRACT

Endogenous lipid stores are thought to be involved in the mechanism whereby the beta-cell adapts its secretory capacity in obesity and diabetes. In addition, hormone-sensitive lipase (HSL) is expressed in beta-cells and may provide fatty acids necessary for the generation of coupling factors linking glucose metabolism to insulin release. We have recently created HSL-deficient mice that were used to directly assess the role of HSL in insulin secretion and action. HSL(-/-) mice were normoglycemic and normoinsulinemic under basal conditions, but showed an approximately 30% reduction of circulating free fatty acids (FFAs) with respect to control and heterozygous animals after an overnight fast. An intraperitoneal glucose tolerance test revealed that HSL-null mice were glucose-intolerant and displayed a lack of a rise in plasma insulin after a glucose challenge. Examination of plasma glucose during an insulin tolerance test suggested that HSL-null mice were insulin-resistant, because plasma glucose was barely lowered after the injection of insulin. Freshly isolated islets from HSL-deficient mice displayed elevated secretion at low (3 mmol/l) glucose, failed to release insulin in response to high (20 mmol/l) glucose, but had a normal secretion when challenged with elevated KCl. The phenotype of heterozygous mice with respect to the measured parameters in vitro was similar to that of wild type. Finally, the islet triglyceride content of HSL(-/-) mice was 2-2.5 fold that in HSL(-/+) and HSL(+/+) animals. The results demonstrate an important role of HSL and endogenous beta-cell lipolysis in the coupling mechanism of glucose-stimulated insulin secretion. The data also provide direct support for the concept that some lipid molecule(s), such as FFAs, fatty acyl-CoA or their derivatives, are implicated in beta-cell glucose signaling.


Subject(s)
Glucose/physiology , Insulin/metabolism , Sterol Esterase/physiology , Animals , Blood Glucose/metabolism , Glucose/pharmacology , Glucose Intolerance/etiology , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Insulin/pharmacology , Insulin Secretion , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Reference Values , Sterol Esterase/deficiency , Sterol Esterase/genetics , Triglycerides/metabolism
14.
Diabetes ; 50(4): 803-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11289045

ABSTRACT

The mechanism by which long-term exposure of the beta-cell to elevated concentrations of fatty acid alters glucose-induced insulin secretion has been examined. Exposure of INS-1 beta-cells to 0.4 mmol/l oleate for 72 h increased basal insulin secretion and decreased insulin release in response to high glucose, but not in response to agents acting at the level of the K(ATP) channel (tolbutamide) or beyond (elevated KCl). This also suppressed the glucose-induced increase in the cellular ATP-to-ADP ratio. The depolarization of the plasma membrane promoted by glucose was decreased after oleate exposure, whereas the response to KCl was unchanged. Cells exposed to free fatty acids displayed a lower mitochondrial membrane potential and a decreased glucose-induced hyperpolarization. The possible implication of uncoupling protein (UCP)-2 in the altered secretory response was examined by measuring UCP2 gene expression after chronic exposure of the cells to fatty acids. UCP2 mRNA and protein were increased twofold by oleate. Palmitate and the nonoxidizable fatty acid bromopalmitate had similar effects on UCP2 mRNA, suggesting that UCP2 gene induction by fatty acids does not require their metabolism. The data are compatible with a role of UCP2 and partial mitochondrial uncoupling in the decreased secretory response to glucose observed after chronic exposure of the beta-cell to elevated fatty acids, and suggest that the expression and/or activity of the protein may modulate insulin secretion in response to glucose.


Subject(s)
Fatty Acids/metabolism , Glucose/physiology , Insulin/metabolism , Membrane Transport Proteins , Mitochondrial Proteins , Proteins/physiology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Blood Physiological Phenomena , Cell Line , Fatty Acids/pharmacology , Gene Expression/drug effects , Insulin Antagonists/pharmacology , Insulin Secretion , Intracellular Membranes/physiology , Ion Channels , Islets of Langerhans/metabolism , Membrane Potentials/drug effects , Mitochondria/physiology , Oleic Acid/pharmacology , Proteins/genetics , Time Factors , Uncoupling Protein 2
15.
Am J Physiol Endocrinol Metab ; 280(2): E238-47, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158926

ABSTRACT

Regional differences in free fatty acid (FFA) handling contribute to diseases associated with particular fat distributions. As cultured rat preadipocytes became differentiated, FFA transfer into preadipocytes increased and was more rapid in single perirenal than in epididymal cells matched for lipid content. Uptake by human omental preadipocytes was greater than uptake by abdominal subcutaneous preadipocytes. Adipose-specific fatty acid binding protein (aP2) and keratinocyte lipid binding protein abundance was higher in differentiated rat perirenal than in epididymal preadipocytes. This interdepot difference in preadipocyte aP2 expression was reflected in fat tissue in older animals. Carnitine palmitoyltransferase 1 activity increased during differentiation and was higher in perirenal than in epididymal preadipocytes, particularly the muscle isoform. Long-chain acyl-CoA levels were higher in perirenal than in epididymal preadipocytes and isolated fat cells. These data are consistent with interdepot differences in fatty acid flux ensuing from differences in fatty acid binding proteins and enzymes of fat metabolism. Heterogeneity among depots results, in part, from distinct intrinsic characteristics of adipose cells. Different depots are effectively separate miniorgans.


Subject(s)
Adipocytes/metabolism , Fatty Acids, Nonesterified/metabolism , Neoplasm Proteins , Nerve Tissue Proteins , Stem Cells/metabolism , Tumor Suppressor Proteins , Acyl Coenzyme A/metabolism , Adult , Animals , Carnitine O-Palmitoyltransferase/metabolism , Carrier Proteins/metabolism , Cells, Cultured , Epididymis , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Female , Humans , Kidney , Male , Middle Aged , Omentum/cytology , Rats , Rats, Inbred F344 , Substrate Specificity
16.
Diabetes ; 49(12): 2012-20, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11118002

ABSTRACT

Administration of dehydroepiandrosterone (DHEA), or its sulfated form (DHEAS), controls hyperglycemia in diabetic rodents without directly altering insulin sensitivity. We show that DHEAS enhanced glucose-stimulated insulin secretion when administered in vivo to rats or in vitro to beta-cell lines, without changing cellular insulin content. Insulin secretion increased from 3 days of steroid exposure in vitro, suggesting that DHEAS did not directly activate the secretory processes. DHEAS selectively increased the beta-cell mRNA expression of acyl CoA synthetase-2 and peroxisomal acyl CoA oxidase in a time-dependent manner. Although DHEAS is a peroxisomal proliferator, it did not alter the mRNA expression of peroxisomal proliferator-activated receptor (PPAR) alpha or beta, or enhance the activity of transfected PPAR alpha, beta, or gamma in vitro. Thus, DHEAS directly affected the beta-cell to enhance glucose-stimulated insulin secretion and increased the mRNA expression of specific beta-cell mitochondrial and peroxisomal lipid metabolic enzymes. This effect of DHEAS on insulin secretion may contribute to the amelioration of hyperglycemia seen in various rodent models of diabetes.


Subject(s)
Dehydroepiandrosterone Sulfate/pharmacology , Gene Expression Regulation/drug effects , Glucose/physiology , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Acyl-CoA Oxidase , Animals , Cell Line , Coenzyme A Ligases/genetics , Insulin Secretion , Male , Mitochondrial Proteins , Oxidoreductases/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
17.
Cancer Res ; 60(22): 6353-8, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11103797

ABSTRACT

Epidemiological studies and experiments using animal models and cultured breast cancer cells have suggested that a high intake of dietary fat could increase breast cancer risk. Little is known about the biochemical pathways by which various free fatty acids (FFAs) influence breast cancer cell proliferation and apoptosis. The present study was designed to investigate the effects of the two most abundant circulating FFAs, oleate and palmitate, on established human breast cancer cell lines after a short period of serum starvation. The unsaturated FFA oleate (C:18:1) stimulated cell proliferation, whereas the saturated FFA palmitate (C:16) dose dependently inhibited it. The half maximal effective concentrations of oleate and palmitate in the presence of albumin were 5 and 25 microM, respectively. The growth-inhibitory effect of palmitate in MDA-MB-231 cells was related to the induction of apoptosis as indicated by morphological and biochemical criteria. Moreover, oleate protected cells against the proapoptotic action of palmitate. Oleate and palmitate increased and decreased phophatidylinositol 3-kinase (PI3-K) activity, respectively, and the actions of the two FFAs on the enzyme were antagonistic. The PI3-K inhibitors wortmannin and LY294002 completely blocked the proliferative action of oleate. 2-Bromopalmitate, a nonmetabolizable analogue, did not affect MDA-MB-231 cell proliferation, suggesting that palmitate must be metabolized to exert its effect. Thus, various types of fatty acids are not equivalent with respect to their actions on breast cancer cell proliferation and apoptosis. The results support the concept that PI3-K is implicated in the control of breast cancer cell growth by FFAs and that PI3-K may provide a link between fat and cancer. The data are also consistent with the view that the type of FFA and their ratios in the diet in addition to the total amount of fat influence mammary carcinogenesis.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Oleic Acid/pharmacology , Palmitic Acid/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Cell Division/drug effects , Culture Media, Serum-Free , Drug Interactions , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Palmitic Acid/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Tumor Cells, Cultured/drug effects
18.
Semin Cell Dev Biol ; 11(4): 267-75, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10966860

ABSTRACT

Glucose-induced insulin secretion is pulsatile. Glucose metabolism generates oscillations in the ATP/ADP ratio which lead to opening and closing of ATP-sensitive K(+)-channels producing subsequent oscillations in membrane potential, cytoplasmic calcium and insulin release. Metabolic signals derived from glucose can also stimulate insulin release independent of their effects on ATP-sensitive K(+)-channels. The ATP/ADP ratio may mediate both ATP-sensitive K(+)-channel-dependent and -independent pathways of secretion. Glucose metabolism also results in an increase in long-chain acyl-CoA, which is proposed to act as an effector molecule in the beta -cell. Long-chain acyl-CoA has a variety of effects in the beta -cell that may effect insulin secretion including opening ATP-sensitive K(+)-channels, activating endoplasmic reticulum Ca(2+)-ATPases and stimulating classical protein kinase C activity. In addition to stimulating insulin release, nutrients also effect gene expression, protein synthesis and beta -cell proliferation. Gene expression is effected by nutrient induction of a variety of immediate early response genes. Glucose stimulates proinsulin biosynthesis both at the translational and transcriptional level. beta -cell proliferation, as a result of insulin-like growth factor and growth hormone mitogenic pathways, is also glucose dependent. Thus, many beta -cell functions in addition to secretion are controlled by nutrient metabolism.


Subject(s)
Islets of Langerhans/physiology , Acyl Coenzyme A/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Calcium/metabolism , Cell Division , Gene Expression Regulation , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Models, Biological , Nucleotides/metabolism
19.
J Biol Chem ; 275(46): 35799-806, 2000 Nov 17.
Article in English | MEDLINE | ID: mdl-10967113

ABSTRACT

To better understand the action of glucose on fatty acid metabolism in the beta-cell and the link between chronically elevated glucose or fatty acids and beta-cell decompensation in adipogenic diabetes, we investigated whether glucose regulates peroxisomal proliferator-activated receptor (PPAR) gene expression in the beta-cell. Islets or INS(832/13) beta-cells exposed to high glucose show a 60-80% reduction in PPARalpha mRNA expression. Oleate, either in the absence or presence of glucose, has no effect. The action of glucose is dose-dependent in the 6-20 mm range and maximal after 6 h. Glucose also causes quantitatively similar reductions in PPARalpha protein and DNA binding activity of this transcription factor. The effect of glucose is blocked by the glucokinase inhibitor mannoheptulose, is partially mimicked by 2-deoxyglucose, and is not blocked by the 3-O-methyl or the 6-deoxy analogues of the sugar that are not phosphorylated. Chronic elevated glucose reduces the expression levels of the PPAR target genes, uncoupling protein 2 and acyl-CoA oxidase, which are involved in fat oxidation and lipid detoxification. A 3-day exposure of INS-1 cells to elevated glucose results in a permanent rise in malonyl-CoA, the inhibition of fat oxidation, and the promotion of fatty acid esterification processes and causes elevated insulin secretion at low glucose. The results suggest that a reduction in PPARalpha gene expression together with a rise in malonyl-CoA plays a role in the coordinated adaptation of beta-cell glucose and lipid metabolism to hyperglycemia and may be implicated in the mechanism of beta-cell "glucolipotoxicity."


Subject(s)
Down-Regulation/drug effects , Glucose/pharmacology , Islets of Langerhans/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Glucose/analogs & derivatives , Glucose/metabolism , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Kinetics , Malonyl Coenzyme A/metabolism , Models, Biological , Oleic Acid/metabolism , Oleic Acid/pharmacology , Oxidation-Reduction/drug effects , Palmitic Acid/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Response Elements/drug effects , Transcription Factors/metabolism , Triglycerides/metabolism
20.
Biochem J ; 350 Pt 2: 599-608, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10947976

ABSTRACT

In the liver, malonyl-CoA is central to many cellular processes, including both fatty acid biosynthesis and oxidation. Malonyl-CoA decarboxylase (MCD) is involved in the control of cellular malonyl-CoA levels, and functions to decarboxylate malonyl-CoA to acetyl-CoA. MCD may play an essential role in regulating energy utilization in the liver by regulating malonyl-CoA levels in response to various nutritional or pathological states. The purpose of the present study was to investigate the role of liver MCD in the regulation of fatty acid oxidation in situations where lipid metabolism is altered. A single MCD enzyme of molecular mass 50.7 kDa was purified from rat liver using a sequential column chromatography procedure and the cDNA was subsequently cloned and sequenced. The liver MCD cDNA was identical to rat pancreatic beta-cell MCD cDNA, and contained two potential translational start sites, producing proteins of 50.7 kDa and 54.7 kDa. Western blot analysis using polyclonal antibodies generated against rat liver MCD showed that the 50.7 kDa isoform of MCD is most abundant in heart and liver, and of relatively low abundance in skeletal muscle (despite elevated MCD transcript levels in skeletal muscle). Tissue distribution experiments demonstrated that the pancreas is the only rat tissue so far identified that contains both the 50.7 kDa and 54. 7 kDa isoforms of MCD. In addition, transfection of the full-length rat liver MCD cDNA into COS cells produced two isoforms of MCD. This indicated either that both initiating methionines are functionally active, generating two proteins, or that the 54.7 kDa isoform is the only MCD protein translated and removal of the putative mitochondrial targeting pre-sequence generates a protein of approx. 50.7 kDa in size. To address this, we transiently transfected a mutated MCD expression plasmid (second ATG to GCG) into COS-7 cells and performed Western blot analysis using our anti-MCD antibody. Western blot analysis revealed that two isoforms of MCD were still present, demonstrating that the second ATG may not be responsible for translation of the 50.7 kDa isoform of MCD. These data also suggest that the smaller isoform of MCD may originate from intracellular processing. To ascertain the functional role of the 50. 7 kDa isoform of rat liver MCD, we measured liver MCD activity and expression in rats subjected to conditions which are known to alter fatty acid metabolism. The activity of MCD was significantly elevated under conditions in which hepatic fatty acid oxidation is known to increase, such as streptozotocin-induced diabetes or following a 48 h fast. A 2-fold increase in expression was observed in the streptozotocin-diabetic rats compared with control rats. In addition, MCD activity was shown to be enhanced by alkaline phosphatase treatment, suggesting phosphorylation-related control of the enzyme. Taken together, our data demonstrate that rat liver expresses a 50.7 kDa form of MCD which does not originate from the second methionine of the cDNA sequence. This MCD is regulated by at least two mechanisms (only one of which is phosphorylation), and its activity and expression are increased under conditions where fatty acid oxidation increases.


Subject(s)
Carboxy-Lyases/chemistry , Carboxy-Lyases/physiology , Fatty Acids/metabolism , Liver/enzymology , Oxygen/metabolism , Alkaline Phosphatase/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Blood Glucose/metabolism , Blotting, Western , COS Cells , Chromatography, Agarose , Cloning, Molecular , DNA, Complementary/metabolism , Diabetes Mellitus, Experimental/metabolism , Fatty Acids/blood , Food Deprivation , Insulin/blood , Liver/metabolism , Male , Methionine/chemistry , Molecular Sequence Data , Myocardium/metabolism , Phosphorylation , Protein Biosynthesis , Protein Isoforms , Rats , Rats, Sprague-Dawley , Sequence Analysis, DNA , Streptozocin , Tissue Distribution , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...