Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 292(5): 1925-1933, 2017 02 03.
Article in English | MEDLINE | ID: mdl-27994053

ABSTRACT

Apelin-36 was discovered as the endogenous ligand for the previously orphan receptor APJ. Apelin-36 has been linked to two major types of biological activities: cardiovascular (stimulation of cardiac contractility and suppression of blood pressure) and metabolic (improving glucose homeostasis and lowering body weight). It has been assumed that both of these activities are modulated through APJ. Here, we demonstrate that the metabolic activity of apelin-36 can be separated from canonical APJ activation. We developed a series of apelin-36 variants in which evolutionarily conserved residues were mutated, and evaluated their ability to modulate glucose homeostasis and body weight in chronic mouse models. We found that apelin-36(L28A) retains full metabolic activity, but is 100-fold impaired in its ability to activate APJ. In contrast to its full metabolic activity, apelin-36(L28A) lost the ability to suppress blood pressure in spontaneously hypertensive rats (SHR). We took advantage of these findings to develop a longer-acting variant of apelin-36 that could modulate glucose homeostasis without impacting blood pressure (or activating APJ). Apelin-36-[L28C(30kDa-PEG)] is 10,000-fold less potent than apelin-36 at activating the APJ receptor but retains its ability to significantly lower blood glucose and improve glucose tolerance in diet-induced obese mice. Apelin-36-[L28C(30kDa-PEG)] provides a starting point for the development of diabetes therapeutics that are devoid of the blood pressure effects associated with canonical APJ activation.


Subject(s)
Adipokines/pharmacology , Blood Glucose/metabolism , Body Weight/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Animals , Apelin , Apelin Receptors , Blood Pressure/drug effects , Mice , Rats , Rats, Inbred SHR
2.
Hepatology ; 65(3): 950-968, 2017 03.
Article in English | MEDLINE | ID: mdl-27880981

ABSTRACT

Because nonalcoholic steatohepatitis (NASH) is associated with impaired liver regeneration, we investigated the effects of G49, a dual glucagon-like peptide-1/glucagon receptor agonist, on NASH and hepatic regeneration. C57Bl/6 mice fed chow or a methionine and choline-deficient (MCD) diet for 1 week were divided into 4 groups: control (chow diet), MCD diet, chow diet plus G49, and M+G49 (MCD diet plus G49). Mice fed a high-fat diet (HFD) for 10 weeks were divided into groups: HFD and H+G49 (HFD plus G49). Following 2 (MCD groups) or 3 (HFD groups) weeks of treatment with G49, partial hepatectomy (PH) was performed, and all mice were maintained on the same treatment schedule for 2 additional weeks. Analysis of liver function, hepatic regeneration, and comprehensive genomic and metabolic profiling were conducted. NASH was ameliorated in the M+G49 group, manifested by reduced inflammation, steatosis, oxidative stress, and apoptosis and increased mitochondrial biogenesis. G49 treatment was also associated with replenishment of intrahepatic glucose due to enhanced gluconeogenesis and reduced glucose use through the pentose phosphate cycle and oxidative metabolism. Following PH, G49 treatment increased survival, restored the cytokine-mediated priming phase, and enhanced the proliferative capacity and hepatic regeneration ratio in mice on the MCD diet. NASH markers remained decreased in M+G49 mice after PH, and glucose use was shifted to the pentose phosphate cycle and oxidative metabolism. G49 administered immediately after PH was also effective at alleviating the pathological changes induced by the MCD diet. Benefits in terms of liver regeneration were also found in mice fed HFD and treated with G49. CONCLUSION: Dual-acting glucagon-like peptide-1/glucagon receptor agonists such as G49 represent a novel therapeutic approach for patients with NASH and particularly those requiring PH. (Hepatology 2017;65:950-968).


Subject(s)
Glucagon-Like Peptide 1/antagonists & inhibitors , Liver Regeneration/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Receptors, Glucagon/antagonists & inhibitors , Animals , Biopsy, Needle , Disease Models, Animal , Glucagon-Like Peptide 1/pharmacology , Humans , Immunohistochemistry , Lipid Peroxidation , Liver Regeneration/physiology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress , Random Allocation , Receptors, Glucagon/administration & dosage , Treatment Outcome
3.
Nat Commun ; 12(1): 6951, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34845204

ABSTRACT

To improve the power of mediation in high-throughput studies, here we introduce High-throughput mediation analysis (Hitman), which accounts for direction of mediation and applies empirical Bayesian linear modeling. We apply Hitman in a retrospective, exploratory analysis of the SLIMM-T2D clinical trial in which participants with type 2 diabetes were randomized to Roux-en-Y gastric bypass (RYGB) or nonsurgical diabetes/weight management, and fasting plasma proteome and metabolome were assayed up to 3 years. RYGB caused greater improvement in HbA1c, which was mediated by growth hormone receptor (GHR). GHR's mediation is more significant than clinical mediators, including BMI. GHR decreases at 3 months postoperatively alongside increased insulin-like growth factor binding proteins IGFBP1/BP2; plasma GH increased at 1 year. Experimental validation indicates (1) hepatic GHR expression decreases in post-bariatric rats; (2) GHR knockdown in primary hepatocytes decreases gluconeogenic gene expression and glucose production. Thus, RYGB may induce resistance to diabetogenic effects of GH signaling.Trial Registration: Clinicaltrials.gov NCT01073020.


Subject(s)
Diabetes Mellitus, Type 2/blood , Gastric Bypass , Liver/metabolism , Metabolome , Obesity/blood , Proteome , Animals , Biomarkers/blood , Blood Glucose/metabolism , Body Mass Index , Carrier Proteins/blood , Carrier Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/surgery , Dipeptidases/blood , Dipeptidases/genetics , Fasting/physiology , Gene Expression Regulation , Glycated Hemoglobin/genetics , Glycated Hemoglobin/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Human Growth Hormone/blood , Human Growth Hormone/genetics , Humans , Insulin-Like Growth Factor Binding Protein 1/blood , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 2/blood , Insulin-Like Growth Factor Binding Protein 2/genetics , Liver/pathology , Obesity/genetics , Obesity/pathology , Obesity/surgery , Primary Cell Culture , Rats , Retrospective Studies
4.
J Cell Physiol ; 225(1): 214-22, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20506299

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of insulin and tyrosine kinase growth factor signaling. We have recently demonstrated that PTP1B deficiency increases GLUT2/insulin receptor (IR) A complexes and glucose uptake in suckling, but not adult, primary hepatocytes. Herein we have investigated intrahepatic glucose utilization in 3-5 days old wild-type and PTP1B(-/-) mice. PTP1B deficiency decreased glycogen, lactate, and pyruvate content in the livers from suckling mice. Conversely, the activity of glucose 6-phosphate dehydrogenase (G6PD), the rate limiting enzyme of the pentose phosphate cycle (PPC) which provides substrates for DNA synthesis, was enhanced in the liver of PTP1B(-/-) animals. Liver weight, liver-to-body mass ratio, DNA content, and PCNA expression were increased in PTP1B(-/-) suckling mice compared to the wild-type controls. At the molecular level, STAT 5B phosphorylation, IGF-I mRNA, and protein levels as well as IGF-IR tyrosine phosphorylation were increased in the livers of PTP1B-deficient neonates. Unexpectedly, hepatic and serum triglycerides (TG) were increased by PTP1B deficiency, although the expression of lipogenic enzymes remained as in the wild-type controls. However, the analysis of milk composition revealed higher TG content in lactating females lacking PTP1B. The effects of PTP1B deficiency on G6PD activity, STAT 5B/IGF-I/IGF-IR axis, PCNA expression and liver growth during suckling were maintained by transferring PTP1B(-/-) embryos (PTP1B(-/-T)) to a wild-type female. Conversely, PTP1B(-/-T) mice did not show hepatic fat accumulation. In conclusion, the present study suggests that PTP1B plays a unique role in the control of the physiological liver development after birth.


Subject(s)
Animals, Suckling , Insulin-Like Growth Factor I/metabolism , Liver/growth & development , Liver/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Animals , Female , Glucose/metabolism , Insulin-Like Growth Factor I/genetics , Lactation/physiology , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proliferating Cell Nuclear Antigen/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Triglycerides/metabolism
5.
Nat Metab ; 2(5): 413-431, 2020 05.
Article in English | MEDLINE | ID: mdl-32478287

ABSTRACT

Non-alcoholic fatty liver disease and steatohepatitis are highly associated with obesity and type 2 diabetes mellitus. Cotadutide, a GLP-1R/GcgR agonist, was shown to reduce blood glycemia, body weight and hepatic steatosis in patients with T2DM. Here, we demonstrate that the effects of Cotadutide to reduce body weight, food intake and improve glucose control are predominantly mediated through the GLP-1 signaling, while, its action on the liver to reduce lipid content, drive glycogen flux and improve mitochondrial turnover and function are directly mediated through Gcg signaling. This was confirmed by the identification of phosphorylation sites on key lipogenic and glucose metabolism enzymes in liver of mice treated with Cotadutide. Complementary metabolomic and transcriptomic analyses implicated lipogenic, fibrotic and inflammatory pathways, which are consistent with a unique therapeutic contribution of GcgR agonism by Cotadutide in vivo. Significantly, Cotadutide also alleviated fibrosis to a greater extent than Liraglutide or Obeticholic acid (OCA), despite adjusting dose to achieve similar weight loss in 2 preclinical mouse models of NASH. Thus Cotadutide, via direct hepatic (GcgR) and extra-hepatic (GLP-1R) effects, exerts multi-factorial improvement in liver function and is a promising therapeutic option for the treatment of steatohepatitis.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Lipogenesis/drug effects , Liver Cirrhosis/drug therapy , Mitochondria/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Peptides/therapeutic use , Animals , Blood Glucose/metabolism , Body Weight , Diabetes Mellitus, Type 2/complications , Glucagon-Like Peptide-1 Receptor/genetics , Glycogen/metabolism , Liver/drug effects , Liver/enzymology , Liver/metabolism , Liver Cirrhosis/metabolism , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Proteomics
6.
Diabetes ; 56(2): 404-13, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17259385

ABSTRACT

Protein-tyrosine phosphatase (PTP)1B is a negative regulator of insulin signaling and a therapeutic target for type 2 diabetes. In this study, we have assessed the role of PTP1B in the insulin sensitivity of skeletal muscle under physiological and insulin-resistant conditions. Immortalized myocytes have been generated from PTP1B-deficient and wild-type neonatal mice. PTP1B(-/-) myocytes showed enhanced insulin-dependent activation of insulin receptor autophosphorylation and downstream signaling (tyrosine phosphorylation of insulin receptor substrate [IRS]-1 and IRS-2, activation of phosphatidylinositol 3-kinase, and serine phosphorylation of AKT), compared with wild-type cells. Accordingly, PTP1B(-/-) myocytes displayed higher insulin-dependent stimulation of glucose uptake and GLUT4 translocation to the plasma membrane than wild-type cells. Treatment with tumor necrosis factor-alpha (TNF-alpha) induced insulin resistance on glucose uptake, impaired insulin signaling, and increased PTP1B activity in wild-type cells. Conversely, the lack of PTP1B confers protection against insulin resistance by TNF-alpha in myocyte cell lines and in adult male mice. Wild-type mice treated with TNF-alpha developed a pronounced hyperglycemia along the glucose tolerance test, accompanied by an impaired insulin signaling and increased PTP1B activity in muscle. However, mice lacking PTP1B maintained a rapid clearance of glucose and insulin sensitivity and displayed normal muscle insulin signaling regardless the presence of TNF-alpha.


Subject(s)
Glucose Transporter Type 4/drug effects , Insulin Resistance/physiology , Insulin/metabolism , Muscle Cells/metabolism , Protein Tyrosine Phosphatases/deficiency , Tumor Necrosis Factor-alpha/pharmacology , Animals , Animals, Newborn , Cell Line , Disease Models, Animal , Genetic Vectors , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Male , Mice , Mice, Knockout , Muscle, Skeletal/cytology , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/metabolism , Receptor, Insulin/drug effects , Signal Transduction , Transfection
7.
Cell Metab ; 27(2): 461-469.e6, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29233536

ABSTRACT

Ghrelin, an appetite-stimulatory hormone secreted by the stomach, was discovered as a ligand for the growth hormone secretagogue receptor (GHSR). Through GHSR, ghrelin stimulates growth hormone (GH) secretion, a function that evolved to protect against starvation-induced hypoglycemia. Though the biology mediated by ghrelin has been described in great detail, regulation of ghrelin action is poorly understood. Here, we report the discovery of liver-expressed antimicrobial peptide 2 (LEAP2) as an endogenous antagonist of GHSR. LEAP2 is produced in the liver and small intestine, and its secretion is suppressed by fasting. LEAP2 fully inhibits GHSR activation by ghrelin and blocks the major effects of ghrelin inĀ vivo, including food intake, GHĀ release, and maintenance of viable glucose levelsĀ during chronic caloric restriction. In contrast, neutralizing antibodies that block endogenous LEAP2 function enhance ghrelin action inĀ vivo. OurĀ findings reveal a mechanism for fine-tuning ghrelin action in response to changing environmental conditions.


Subject(s)
Hepcidins/metabolism , Receptors, Ghrelin/antagonists & inhibitors , Animals , Bariatric Surgery , Caloric Restriction , Eating , Fasting , Female , Ghrelin/antagonists & inhibitors , Ghrelin/metabolism , Growth Hormone/metabolism , Humans , Intestine, Small/metabolism , Liver/metabolism , Male , Mice , Protein Binding , Rats , Receptors, Ghrelin/metabolism
8.
Sci Rep ; 8(1): 16461, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30405191

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is associated with post-operative liver failure (PLF) and impaired liver regeneration. We investigated the effects of a glucagon-like peptide-1 (GLP-1) receptor agonist on NAFLD, PLF and liver regeneration in mice fed chow diet or methionine/choline-deficient diet (MCD) or high fat diet (HFD). Fc-GLP-1 decreased transaminases, reduced intrahepatic triglyceridesĀ (TG) and improved MCD-induced liver dysfuction. Macrophage/Kupffer cell-related markers were also reduced although Fc-GLP-1 increased expression of genes related to natural killer (NK), cytotoxic T lymphocytes and hepatic stellate cell (HSC) activation. After partial hepatectomy (PH), survival rates increased in mice receiving Fc-GLP-1 on chow or MCD diet. However, the benefit of Fc-GLP-1 on NASH-like features was attenuated 2 weeks post-PH and liver mass restoration was not improved. At this time-period, markers of NK cells and cytotoxic T lymphocytes were further elevated in Fc-GLP-1Ā treated mice. Increased HSCĀ related gene expression in livers was observed together with decreased retinyl ester content and increased retinal and retinoic acid, reflecting HSC activation. Similar effects were found in mice fed HFD receiving Fc-GLP-1. Our results shed light on the differential effects of a long-acting GLP-1R agonist in improving NAFLD and PLF, but not enhancing liver regeneration in mice.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Hepatectomy , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Biomarkers , Biopsy , Disease Models, Animal , Gene Expression , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Immunohistochemistry , Inflammation Mediators , Liver/immunology , Liver/metabolism , Liver/pathology , Liver/surgery , Liver Regeneration , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology
9.
Sci Rep ; 8(1): 17545, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30510163

ABSTRACT

Type 2 diabetes (T2D) is a complex and progressive disease requiring polypharmacy to manage hyperglycaemia and cardiovascular risk factors. However, most patients do not achieve combined treatment goals. To address this therapeutic gap, we have developed MEDI4166, a novel glucagon-like peptide-1 (GLP-1) receptor agonist peptide fused to a proprotein convertase subtilisin/kexin type 9 (PCSK9) neutralising antibody that allows for glycaemic control and low-density lipoprotein cholesterol (LDL-C) lowering in a single molecule. The fusion has been engineered to deliver sustained peptide activity in vivo in combination with reduced potency, to manage GLP-1 driven adverse effects at high dose, and a favourable manufacturability profile. MEDI4166 showed robust and sustained LDL-C lowering in cynomolgus monkeys and exhibited the anticipated GLP-1 effects in T2D mouse models. We believe MEDI4166 is a novel molecule combining long acting agonist peptide and neutralising antibody activities to deliver a unique pharmacology profile for the management of T2D.


Subject(s)
Antibodies, Monoclonal , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Glucagon-Like Peptide 1 , Hypoglycemic Agents , PCSK9 Inhibitors , Recombinant Fusion Proteins , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , CHO Cells , Cricetulus , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Hep G2 Cells , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Macaca fascicularis , Male , Mice , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology
10.
Endocrinology ; 148(2): 594-608, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17068137

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of insulin signaling and a therapeutic target for type 2 diabetes. The purpose of this study was to evaluate the differences in insulin sensitivity between neonate and adult hepatocytes lacking PTP1B. Immortalized neonatal hepatocytes and primary neonatal and adult hepatocytes have been generated from PTP1B(-/-) and wild-type mice. PTP1B deficiency in immortalized neonatal hepatocytes prolonged insulin-induced tyrosine phosphorylation of the insulin receptor (IR) and IR substrates (IRS) -1, -2 compared with wild-type control cells. Endogenous IR and IRS-2 were down-regulated, whereas IRS-1 was up-regulated in PTP1B(-/-) neonatal hepatocytes and livers of PTP1B(-/-) neonates. Insulin-induced activation of phosphatidylinositol 3-kinase/Akt pathway was prolonged in PTP1B(-/-) immortalized neonatal hepatocytes. However, insulin sensitivity was comparable to wild-type hepatocytes. Rescue of PTP1B in deficient cells suppressed the prolonged insulin signaling, whereas RNA interference in wild-type cells promoted prolonged signaling. In primary neonatal PTP1B(-/-) hepatocytes, insulin prolonged the inhibition of gluconeogenic mRNAs, but the sensitivity to this inhibition was similar to wild-type cells. By contrast, in adult PTP1B-deficient livers, p85alpha was down-regulated compared with the wild type. Moreover, primary hepatocytes from adult PTP1B(-/-) mice displayed enhanced Akt phosphorylation and a more pronounced inhibition of gluconeogenic mRNAs than wild-type cells. Hepatic insulin sensitivity due to PTP1B deficiency is acquired through postnatal development. Thus, changes in IR and IRS-2 expression and in the balance between regulatory and catalytic subunits of phosphatidylinositol 3-kinase are necessary to achieve insulin sensitization in adult PTP1B(-/-) hepatocytes.


Subject(s)
Aging/physiology , Hepatocytes/physiology , Insulin Resistance , Insulin/physiology , Protein Tyrosine Phosphatases/deficiency , Animals , Animals, Newborn , Cell Line, Transformed , Down-Regulation , Gene Expression , Gene Expression Profiling , Gene Transfer Techniques , Gluconeogenesis/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Insulin/metabolism , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , RNA, Small Interfering/pharmacology , Receptor, Insulin/metabolism , Signal Transduction , Time Factors , Tyrosine/metabolism , Up-Regulation
11.
Diabetes ; 66(7): 2007-2018, 2017 07.
Article in English | MEDLINE | ID: mdl-28408435

ABSTRACT

Neurturin (NRTN), a member of the glial-derived neurotrophic factor family, was identified from an embryonic chicken pancreatic cDNA library in a screen for secreted factors. In this study, we assessed the potential antidiabetic activities of NRTN relative to liraglutide, a glucagon-like peptide 1 receptor agonist, in Zucker diabetic fatty (ZDF) rats. Subcutaneous administration of NRTN to 8-week-old male ZDF rats prevented the development of hyperglycemia and improved metabolic parameters similar to liraglutide. NRTN treatment increased pancreatic insulin content and Ɵ-cell mass and prevented deterioration of islet organization. However, unlike liraglutide-treated rats, NRTN-mediated improvements were not associated with reduced body weight or food intake. Acute NRTN treatment did not activate c-Fos expression in key feeding behavior and metabolic centers in ZDF rat brain or directly enhance glucose-stimulated insulin secretion from pancreatic Ɵ-cells. Treating 10-week-old ZDF rats with sustained hyperglycemia with liraglutide resulted in some alleviation of hyperglycemia, whereas NRTN was not as effective despite improving plasma lipids and fasting glucose levels. Interestingly, coadministration of NRTN and liraglutide normalized hyperglycemia and other metabolic parameters, demonstrating that combining therapies with distinct mechanism(s) can alleviate advanced diabetes. This emphasizes that therapeutic combinations can be more effective to manage diabetes in individuals with uncontrolled hyperglycemia.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Liraglutide/pharmacology , Neurturin/pharmacology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Disease Models, Animal , Eating/drug effects , Feeding Behavior/drug effects , Glucagon-Like Peptide-1 Receptor/agonists , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Organ Size , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Zucker
12.
Endocrinology ; 147(6): 2650-6, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16556759

ABSTRACT

Over the past years, RNA interference (RNAi) has exploded as a new approach to manipulate gene expression in mammalian systems. More recently, RNAi has acquired interest as a tool to identify new targets for therapeutic intervention. This review focuses on the current understanding of RNAi biology, how RNAi has been used to study the role of different genes in the pathogenesis of diabetes and obesity, and the use of RNAi screens for the identification of new targets for metabolic diseases. Also reviewed are the in vivo proof of principle experiments that provide the validation of these new targets for the development of RNAi-based therapeutics for diabetes.


Subject(s)
Diabetes Mellitus/therapy , Obesity/therapy , RNA Interference , Adenoviridae/genetics , Agouti-Related Protein , Animals , Apolipoproteins B/genetics , Early Growth Response Protein 1/genetics , Humans , Insulin/physiology , Insulin Receptor Substrate Proteins , Intercellular Signaling Peptides and Proteins , Phosphoproteins/genetics , Proteins/genetics , Signal Transduction , Transcription Factors/genetics
13.
J Med Chem ; 49(12): 3563-80, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16759099

ABSTRACT

The c-Jun N-terminal kinases (JNK-1, -2, and -3) are members of the mitogen activated protein (MAP) kinase family of enzymes. They are activated in response to certain cytokines, as well as by cellular stresses including chemotoxins, peroxides, and irradiation. They have been implicated in the pathology of a variety of different diseases with an inflammatory component including asthma, stroke, Alzheimer's disease, and type 2 diabetes mellitus. In this work, high-throughput screening identified a JNK inhibitor with an excellent kinase selectivity profile. Using X-ray crystallography and biochemical screening to guide our lead optimization, we prepared compounds with inhibitory potencies in the low-double-digit nanomolar range, activity in whole cells, and pharmacokinetics suitable for in vivo use. The new compounds were over 1,000-fold selective for JNK-1 and -2 over other MAP kinases including ERK2, p38alpha, and p38delta and showed little inhibitory activity against a panel of 74 kinases.


Subject(s)
Aminopyridines/chemical synthesis , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Aminopyridines/chemistry , Aminopyridines/pharmacology , Animals , Biological Availability , Cell Line, Tumor , Crystallography, X-Ray , Half-Life , Humans , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 8/chemistry , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Models, Molecular , Phosphorylation , Protein Conformation , Rats , Rats, Sprague-Dawley
14.
Biotechniques ; Suppl: 31-6, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16629385

ABSTRACT

Over the past years RNA interference (RNAi) has exploded as a new approach to manipulate gene expression in mammalian systems. More recently, RNAi has acquired interest as a potential therapeutic strategy. This review focuses on the potential therapeutic use of RNAi for metabolic diseases, the current understanding of RNAi biology, and how RNAi has been utilized to study the role of different genes in the pathogenesis of diabetes and obesity. Also reviewed are the in vivo proof-of-principle experiments that provide the preclinical justification for the development of RNAi-based therapeutics for diabetes and the key challenges that currently limit its application in the clinical setting.


Subject(s)
Gene Targeting/methods , Genetic Therapy/methods , Metabolic Diseases/genetics , Metabolic Diseases/therapy , RNA Interference , RNA, Small Interfering/genetics , Gene Targeting/trends , Genetic Engineering/methods , Genetic Engineering/trends , Genetic Therapy/trends , Humans
15.
Diabetes ; 52(3): 634-41, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12606502

ABSTRACT

Serine and threonine kinases may contribute to insulin resistance and the development of type 2 diabetes. To test the potential for members of the mitogen-activated protein (MAP) kinase family to contribute to type 2 diabetes, we examined basal and insulin-stimulated Erk 1/2, JNK, and p38 phosphorylation in adipocytes isolated from healthy and type 2 diabetic individuals. Maximal insulin stimulation increased the phosphorylation of Erk 1/2 and JNK in healthy control subjects but not type 2 diabetic patients. Insulin stimulation did not increase p38 phosphorylation in either healthy control subjects or type 2 diabetic patients. In type 2 diabetic adipocytes, the basal phosphorylation status of these MAP kinases was significantly elevated and was associated with decreased IRS-1 and GLUT4 in these fat cells. To determine whether MAP kinases were involved in the downregulation of IRS-1 and GLUT4 protein levels, selective inhibitors were used to inhibit these MAP kinases in 3T3-L1 adipocytes treated chronically with insulin. Inhibition of Erk 1/2, JNK, or p38 had no effect on insulin-stimulated reduction of IRS-1 protein levels. However, inhibition of the p38 pathway prevented the insulin-stimulated decrease in GLUT4 protein levels. In summary, type 2 diabetes is associated with an increased basal activation of the MAP kinase family. Furthermore, upregulation of the p38 pathway might contribute to the loss of GLUT4 expression observed in adipose tissue from type 2 diabetic patients.


Subject(s)
Adipocytes/enzymology , Diabetes Mellitus, Type 2/enzymology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Monosaccharide Transport Proteins/analysis , Muscle Proteins , 3T3 Cells , Adult , Animals , Enzyme Activation , Female , Glucose Transporter Type 4 , Humans , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Insulin Resistance , MAP Kinase Kinase 4 , Male , Mice , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphoproteins/analysis , Phosphorylation , p38 Mitogen-Activated Protein Kinases
16.
Diabetes ; 51(8): 2405-11, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12145151

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) has been implicated as a negative regulator of insulin action. Overexpression of PTP1B protein has been observed in insulin-resistant states associated with obesity. Mice lacking a functional PTP1B gene exhibit increased insulin sensitivity and are resistant to weight gain. To investigate the role of PTP1B in adipose tissue from obese animals, hyperglycemic obese (ob/ob) mice were treated with PTP1B antisense oligonucleotide (ISIS-113715). A significant reduction in adiposity correlated with a decrease of PTP1B protein levels in fat. Antisense treatment also influenced the triglyceride content in adipocytes, correlating with a downregulation of genes encoding proteins involved in lipogenesis, such as sterol regulatory element-binding protein 1 and their downstream targets spot14 and fatty acid synthase, as well as other adipogenic genes, lipoprotein lipase, and peroxisome proliferator-activated receptor gamma. In addition, an increase in insulin receptor substrate-2 protein and a differential regulation of the phosphatidylinositol 3-kinase regulatory subunit (p85alpha) isoforms expression were found in fat from antisense-treated animals, although increased insulin sensitivity measured by protein kinase B phosphorylation was not observed. These results demonstrate that PTP1B antisense treatment can modulate fat storage and lipogenesis in adipose tissue and might implicate PTP1B in the enlargement of adipocyte energy stores and development of obesity.


Subject(s)
Adipose Tissue/physiopathology , Gene Expression Regulation/physiology , Insulin/physiology , Lipids/biosynthesis , Oligodeoxyribonucleotides, Antisense/pharmacology , Phosphoproteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Weight Loss/physiology , Adipocytes/metabolism , Adipose Tissue/drug effects , Alternative Splicing , Animals , DNA Primers , Gene Expression Regulation/drug effects , Genetic Variation , Homeostasis , Hyperglycemia/enzymology , Hyperglycemia/genetics , Hyperglycemia/physiopathology , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Isoenzymes/genetics , Mice , Mice, Obese , Phosphatidylinositol 3-Kinases/genetics , Polymerase Chain Reaction , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/genetics , Time Factors , Triglycerides/metabolism
17.
Diabetes ; 52(1): 21-8, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12502489

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of insulin receptor (IR) signal transduction and a drug target for treatment of type 2 diabetes. Using PTP1B antisense oligonucleotides (ASOs), effects of decreased PTP1B levels on insulin signaling in diabetic ob/ob mice were examined. Insulin stimulation, prior to sacrifice, resulted in no significant activation of insulin signaling pathways in livers from ob/ob mice. However, in PTP1B ASO-treated mice, in which PTP1B protein was decreased by 60% in liver, similar stimulation with insulin resulted in increased tyrosine phosphorylation of the IR and IR substrate (IRS)-1 and -2 by threefold, fourfold, and threefold, respectively. IRS-2-associated phosphatidylinositol 3-kinase activity was also increased threefold. Protein kinase B (PKB) serine phosphorylation was increased sevenfold in liver of PTP1B ASO-treated mice upon insulin stimulation, while phosphorylation of PKB substrates, glycogen synthase kinase (GSK)-3alpha and -3beta, was increased more than twofold. Peripheral insulin signaling was increased by PTP1B ASO, as evidenced by increased phosphorylation of PKB in muscle of insulin-stimulated PTP1B ASO-treated animals despite the lack of measurable effects on muscle PTP1B protein. These results indicate that reduction of PTP1B is sufficient to increase insulin-dependent metabolic signaling and improve insulin sensitivity in a diabetic animal model.


Subject(s)
Diabetes Mellitus/physiopathology , Insulin/physiology , Obesity , Protein Serine-Threonine Kinases , Protein Tyrosine Phosphatases/antagonists & inhibitors , Signal Transduction/physiology , Animals , Blood Glucose/analysis , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Insulin/blood , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Liver/enzymology , Mice , Mice, Inbred C57BL/genetics , Muscle, Skeletal/enzymology , Oligonucleotides, Antisense/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, Insulin/metabolism , Serine/metabolism , Tyrosine/metabolism
18.
Curr Opin Investig Drugs ; 6(10): 979-87, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16259218

ABSTRACT

Obesity and insulin resistance are strongly associated with systemic markers of inflammation and endoplasmic reticulum stress. c-Jun N-terminal kinases (JNK) are activated by inflammatory cytokines and have a key role in beta-cell apoptosis and in negative regulation of insulin signaling. JNK1-deficient mice are protected from diet-induced obesity and insulin resistance, while genetically obese mice with targeted mutations in JNK1 are leaner and have reduced insulin and blood glucose levels. These studies validate JNK as a link between inflammation and metabolic diseases and as a promising drug target. This review highlights recent advances in small-molecule inhibitors of JNK that have also been targeted for other diseases with an inflammatory component such as stroke, rheumatoid arthritis, and Alzheimer's and Parkinson's diseases.


Subject(s)
Enzyme Inhibitors/pharmacology , Inflammation/enzymology , Insulin Resistance , Insulin/biosynthesis , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Enzyme Inhibitors/chemistry , Humans , Inflammation/metabolism , Molecular Structure
19.
Metabolism ; 54(7): 895-901, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988698

ABSTRACT

Inhibition of p38, a member of the mitogen-activated protein kinase family, has been shown to prevent the loss of GLUT4 protein expression in insulin-resistant adipocytes without improving insulin receptor substrate 1 (IRS-1) protein levels and presumably insulin signaling. Thus, it was unclear whether p38 inhibitors would have a beneficial effect upon insulin-stimulated glucose uptake. We evaluated the effects of p38 inhibition during the development of insulin resistance upon glucose uptake and components of the insulin signaling pathway to determine the therapeutic value of p38 inhibitors. Treatment with the specific p38 inhibitor, A304000, during the development of insulin resistance increased basal glucose uptake as well as glucose uptake in response to a subsequent insulin stimulation. p38 inhibition increased GLUT1 protein levels and prevented the loss of GLUT4. However, p38 inhibition did not prevent the loss of IRS-1 protein levels or insulin signaling to PKB in insulin-resistant cells. Rapamycin, an inhibitor or mTOR, could partially improve insulin-stimulated glucose uptake through maintaining IRS-1 protein levels. Combined treatment with both A304000 and rapamycin had an additive effect upon glucose uptake. These data indicate that p38 inhibition can enhance glucose uptake through regulating the expression of GLUT1 and 4, but did not prevent the development of insulin resistance.


Subject(s)
Adipocytes/drug effects , Enzyme Inhibitors/pharmacology , Glucose/metabolism , Insulin Resistance , p38 Mitogen-Activated Protein Kinases/metabolism , 3T3-L1 Cells , Adipocytes/enzymology , Adipocytes/metabolism , Animals , Immunoprecipitation , Mice
20.
Metabolism ; 54(11): 1515-23, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16253642

ABSTRACT

In the present study, we investigated the effects of long-term treatment with the endothelin (ET) antagonist atrasentan, an ET(A)-selective antagonist, on whole body glucose metabolism and insulin signaling in a commonly used model of insulin resistance, the Zucker fatty rat. Zucker lean and fatty rats were maintained for 6 weeks on either control or atrasentan-treated water. Euglycemic-hyperinsulinemic clamps (4 mU/kg per minute) were performed at the end of the 6-week treatment on a subset of rats (n=10/treatment). In another subset (n=5/treatment), an insulin tolerance test was performed; liver and muscle tissues were harvested 10 minutes following the challenge for further analysis. Results of the clamps demonstrated that long-term atrasentan treatment significantly increased whole body glucose metabolism in fatty rats compared with vehicle control subjects. Insulin-induced insulin receptor substrate 1 tyrosine and protein kinase B serine phosphorylation were significantly reduced in the liver and muscle of fatty animals compared with their lean littermates. This reduction was overcome with atrasentan treatment in the liver but not in the muscle. There was no difference between lean and fatty animals, however, in insulin receptor substrate 1 and protein kinase B protein expression in the liver and muscle and no effect by atrasentan. In contrast, expression of the regulatory subunit of PI-3 kinase (p85alpha) was significantly increased in the liver but not in the muscle of fatty animals compared with their lean littermates and this was normalized to levels of lean animals with atrasentan treatment. These findings indicate that long-standing ET antagonism improves whole body glucose metabolism in Zucker fatty rats through improvements in insulin signaling in the liver. These results indicate that therapeutic ET antagonism may assist in correcting the insulin-resistant state.


Subject(s)
Endothelin-1/antagonists & inhibitors , Insulin Resistance , Liver/metabolism , Obesity/drug therapy , Obesity/metabolism , Signal Transduction/drug effects , Animals , Atrasentan , Blood Pressure , Body Weight/drug effects , Drinking/drug effects , Endothelin-1/blood , Glucose/metabolism , Glucose Clamp Technique , Insulin/metabolism , Muscle, Skeletal/metabolism , Pyrrolidines/pharmacology , Rats , Rats, Zucker
SELECTION OF CITATIONS
SEARCH DETAIL