1
|
Ford BE, Chachra SS, Alshawi A, Oakley F, Fairclough RJ, Smith DM, Tiniakos D, Agius L. Compromised chronic efficacy of a glucokinase activator AZD1656 in mouse models for common human GCKR variants. Biochem Pharmacol 2024; 229:116499. [PMID: 39173844 DOI: 10.1016/j.bcp.2024.116499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/23/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Glucokinase activators (GKAs) have been developed as blood glucose lowering drugs for type 2 diabetes. Despite good short-term efficacy, several GKAs showed a decline in efficacy chronically during clinical trials. The underlying mechanisms remain incompletely understood. We tested the hypothesis that deficiency in the liver glucokinase regulatory protein (GKRP) as occurs with common human GCKR variants affects chronic GKA efficacy. We used a Gckr-P446L mouse model for the GCKR exonic rs1260326 (P446L) variant and the Gckr-del/wt mouse to model transcriptional deficiency to test for chronic efficacy of the GKA, AZD1656 in GKRP-deficient states. In the Gckr-P446L mouse, the blood glucose lowering efficacy of AZD1656 (3 mg/kg body wt) after 2 weeks was independent of genotype. However after 19 weeks, efficacy was maintained in wild-type but declined in the LL genotype, in conjunction with raised hepatic glucokinase activity and without raised liver lipids. Sustained blood glucose lowering efficacy in wild-type mice was associated with qualitatively similar but more modest changes in the liver transcriptome compared with the P446L genotype, consistent with GKA therapy representing a more modest glucokinase excess than the P446L genotype. Chronic treatment with AZD1656 in the Gckr-del/wt mouse was associated with raised liver triglyceride and hepatocyte microvesicular steatosis. The results show that in mouse models of liver GKRP deficiency in conjunction with functional liver glucokinase excess as occurs in association with common human GCKR variants, GKRP-deficiency predisposes to declining efficacy of the GKA in lowering blood glucose and to GKA induced elevation in liver lipids.
Collapse
Affiliation(s)
- Brian E Ford
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Shruti S Chachra
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Ahmed Alshawi
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Medical Laboratory Technique Department, Kufa Institute, Al-Furat Al-Awsat Technical University, Kufa, Iraq
| | - Fiona Oakley
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Rebecca J Fairclough
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - David M Smith
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Dina Tiniakos
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Dept of Pathology, Aretaieion Hospital Medical School, National and Kapodistrian University of Athens, Greece
| | - Loranne Agius
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
2
|
Raju R, Prabath I, Chandrasekaran I, Varadarajan S. Dorzagliatin: A Breakthrough Glucokinase Activator Coming on Board to Treat Diabetes Mellitus. Cureus 2024; 16:e65708. [PMID: 39211666 PMCID: PMC11361462 DOI: 10.7759/cureus.65708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Dorzagliatin, an innovative dual-acting allosteric oral glucokinase activator that targets glucose homeostasis and insulin resistance, has gained approval for treating type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). The effectiveness of existing antidiabetic treatments in enhancing beta cell (β-cell) activity is restricted. Currently, there are no satisfactory medications available to address the fundamental deficiency in glucose sensing for glucokinase-maturity-onset diabetes of the young (GCK-MODY), which is caused by mutations in the glucokinase gene; researchers have embarked on glucokinase activators. Dorzagliatin enhances the affinity of glucokinase for glucose and glucose-sensing capacity, improves β-cell function, and reduces insulin resistance. Two phase 3 studies, an adjunct trial of dorzagliatin with metformin for T2DM patients and a monotherapy trial for drug-naïve T2DM patients, are key clinical trials that have shown a favorable safety and tolerability profile. They also demonstrated a rapid, sustained reduction in glycated hemoglobin (HbA1c) and a significant decrease in postprandial blood glucose. This review will summarize the substantial clinical evidence supporting the safety and efficacy of dorzagliatin in treating diabetes mellitus (DM) and clarify the molecular mechanisms underlying its action.
Collapse
Affiliation(s)
- Ramya Raju
- Department of Pharmacology, Sri Ramaswamy Memorial (SRM) Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Chengalpattu, IND
| | - Indumathi Prabath
- Department of Pharmacology, Sri Ramaswamy Memorial (SRM) Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Chengalpattu, IND
| | - Indumathi Chandrasekaran
- Department of Pharmacology, Sri Venkateshwaraa Medical College Hospital and Research Centre, Puducherry, IND
| | - Sathyanarayanan Varadarajan
- Department of Pharmacology, Sri Ramaswamy Memorial (SRM) Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Chengalpattu, IND
| |
Collapse
|
3
|
Kaur U, Pathak BK, Meerashahib TJ, Krishna DVV, Chakrabarti SS. Should Glucokinase be Given a Chance in Diabetes Therapeutics? A Clinical-Pharmacological Review of Dorzagliatin and Lessons Learned So Far. Clin Drug Investig 2024; 44:223-250. [PMID: 38460077 DOI: 10.1007/s40261-024-01351-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2024] [Indexed: 03/11/2024]
Abstract
Despite advances in the management of type 2 diabetes mellitus (T2DM), one-third of patients with diabetes do not achieve the desired glycemic goal. Considering this inadequacy, many agents that activate glucokinase have been investigated over the last two decades but were withdrawn before submission for marketing permission. Dorzagliatin is the first glucokinase activator that has been granted approval for T2DM, only in China. As overstimulation of glucokinase is linked with pathophysiological disturbances such as fatty liver and cardiovascular issues and a loss of therapeutic efficacy with time. This review aims to highlight the benefits of glucokinase activators vis-à-vis the risks associated with chronic enzymatic activation. We discuss the multisystem disturbances expected with chronic activation of the enzyme, the lessons learned with glucokinase activators of the past, the major efficacy and safety findings with dorzagliatin and its pharmacological properties, and the status of other glucokinase activators in the pipeline. The approval of dorzagliatin in China was based on the SEED and the DAWN trials, the major pivotal phase III trials that enrolled patients with T2DM with a mean glycosylated hemoglobin of 8.3-8.4%, and a mean age of 53-54.5 years from multiple sites in China. Patients with uncontrolled diabetes, cardiac diseases, organ dysfunction, and a history of severe hypoglycemia were excluded. Both trials had a randomized double-blind placebo-controlled phase of 24 weeks followed by an open-label phase of 28 weeks with dorzagliatin. Drug-naïve patients with T2DM with a disease duration of 11.7 months were enrolled in the SEED trial while the DAWN trial involved patients with T2DM with a mean duration of 71.5 months and receiving background metformin therapy. Compared with placebo, the decline in glycosylated hemoglobin at 24 weeks was more with dorzagliatin with an estimated treatment difference of - 0.57% in the SEED trial and - 0.66% in the DAWN trial. The desired glycosylated hemoglobin (< 7%) was also attained at more than two times higher rates with dorzagliatin. The glycemic improvement was sustained in the SEED trial but decreased over 52 weeks in the DAWN trial. Hyperlipidemia was observed in 12-14% of patients taking dorzagliatin versus 9-11% of patients receiving a placebo. Additional adverse effects noticed over 52 weeks with dorzagliatin included an elevation in liver enzymes, hyperuricemia, hyperlacticacidemia, renal dysfunction, and cardiovascular disturbances. Considering the statistically significant improvement in glycosylated hemoglobin with dorzagliatin in patients with T2DM, the drug may be given a chance in treatment-naïve patients with a shorter disease history. However, with the waning therapeutic efficacy witnessed in patients with long-standing diabetes, which was also one of the potential concerns with previously tested molecules, extended studies involving patients with chronic and uncontrolled diabetes are needed to comment upon the long-term therapeutic performance of dorzagliatin. Likewise, evidence needs to be generated from other countries, patients with organ dysfunction, a history of severe hypoglycemia, cardiac diseases, and elderly patients before extending the use of dorzagliatin. Apart from monitoring lipid profiles, long-term safety studies of dorzagliatin should involve the assessment of serum uric acid, lactate, renal function, liver function, and cardiovascular parameters.
Collapse
Affiliation(s)
- Upinder Kaur
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| | - Bhairav Kumar Pathak
- Department of Pharmacology and Therapeutics, All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| | - Tharik Jalal Meerashahib
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | | | - Sankha Shubhra Chakrabarti
- Department of Geriatric Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
4
|
Patel S, Yan Z, Remedi MS. Intermittent fasting protects β-cell identity and function in a type-2 diabetes model. Metabolism 2024; 153:155813. [PMID: 38307325 PMCID: PMC10985623 DOI: 10.1016/j.metabol.2024.155813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Type 2 diabetes (T2DM) is caused by the interaction of multiple genes and environmental factors. T2DM is characterized by hyperglycemia, insulin secretion deficiency and insulin resistance. Chronic hyperglycemia induces β-cell dysfunction, loss of β-cell mass/identity and β-cell dedifferentiation. Intermittent fasting (IF) a commonly used dietary regimen for weight-loss, also induces metabolic benefits including reduced blood glucose, improved insulin sensitivity, reduced adiposity, inflammation, oxidative-stress and increased fatty-acid oxidation; however, the mechanisms underlying these effects in pancreatic β-cells remain elusive. KK and KKAy, mouse models of polygenic T2DM spontaneously develop hyperglycemia, glucose intolerance, glucosuria, impaired insulin secretion and insulin resistance. To determine the long-term effects of IF on T2DM, 6-weeks old KK and KKAy mice were subjected to IF for 16 weeks. While KKAy mice fed ad-libitum demonstrated severe hyperglycemia (460 mg/dL) at 6 weeks of age, KK mice showed blood glucose levels of 230 mg/dL, but progressively became severely diabetic by 22-weeks. Strikingly, both KK and KKAy mice subjected to IF showed reduced blood glucose and plasma insulin levels, decreased body weight gain, reduced plasma triglycerides and cholesterol, and improved insulin sensitivity. They also demonstrated enhanced expression of the β-cell transcription factors NKX6.1, MAFA and PDX1, and decreased expression of ALDH1a3 suggesting protection from loss of β-cell identity by IF. IF normalized glucose stimulated insulin secretion in islets from KK and KKAy mice, demonstrating improved β-cell function. In addition, hepatic steatosis, gluconeogenesis and inflammation was decreased particularly in KKAy-IF mice, indicating peripheral benefits of IF. These results have important implications as an optional intervention for preservation of β-cell identity and function in T2DM.
Collapse
Affiliation(s)
- Sumit Patel
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO, United States of America
| | - Zihan Yan
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO, United States of America
| | - Maria S Remedi
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO, United States of America; Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO, United States of America; Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO, United States of America.
| |
Collapse
|
5
|
Ren Y, Li L, Wan L, Huang Y, Cao S. Glucokinase as an emerging anti-diabetes target and recent progress in the development of its agonists. J Enzyme Inhib Med Chem 2022; 37:606-615. [PMID: 35067153 PMCID: PMC8788356 DOI: 10.1080/14756366.2021.2025362] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Type 2 diabetes mellitus is a metabolic disorder with complicated pathogenesis, and mono-target therapy often fails to effectively manage the levels of blood glucose. In recent years, the anti-diabetes target glucokinase (GK) has attracted the attention of researchers. It acts as a glucose sensor, triggering counter regulatory responses following a change in glucose levels to aid restoration of normoglycemia. Activation of GK induces glucose metabolism and reduces glucose levels for the treatment of type 2 diabetes. GK agonists (GKA) are a new class of antidiabetic drugs. Among these agents, dorzagliatin is currently being investigated in phase III clinical trials, while PB-201 and AZD-1656 have reached phase II clinical trials. This article describes the mechanism of action of GK in diabetes and of action of GKA at the protein level, and provides a review of the research, trends, and prospects regarding the use of GKA in this setting.
Collapse
Affiliation(s)
- Yixin Ren
- Key Laboratory of Green Chemical Engineering Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, P. R. China
| | - Li Li
- Key Laboratory of Green Chemical Engineering Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, P. R. China
| | - Li Wan
- Key Laboratory of Green Chemical Engineering Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, P. R. China
| | - Yan Huang
- Key Laboratory of Green Chemical Engineering Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, P. R. China
| | - Shuang Cao
- Key Laboratory of Green Chemical Engineering Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, P. R. China
| |
Collapse
|
6
|
Driescher N, Joseph DE, Human VR, Ojuka E, Cour M, Hadebe N, Bester D, Marnewick JL, Lecour S, Lochner A, Essop MF. The impact of sugar-sweetened beverage intake on rat cardiac function. Heliyon 2019; 5:e01357. [PMID: 30949605 PMCID: PMC6429811 DOI: 10.1016/j.heliyon.2019.e01357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 02/20/2019] [Accepted: 03/12/2019] [Indexed: 01/01/2023] Open
Abstract
Aims Although there is evidence linking sugar-sweetened beverage (SSB) intake with the development of cardio-metabolic diseases, the underlying mechanisms remain unclear. The current study therefore evaluated the effects of SSB consumption by establishing a unique in-house in vivo experimental model. Main methods Male Wistar rats were divided into two groups: a) one consuming a popular local SSB (SSB- Jive), and b) a control group (Control-water) for a period of three and six months (n = 6 per group), respectively. Rats were gavaged on a daily basis with an experimental dosage amounting to half a glass per day (in human terms) (SSB vs. water). Cardiac function was assessed at baseline (echocardiography) and following ex vivo ischemia-reperfusion of the isolated perfused working rat heart. Oral glucose tolerance tests and mitochondrial respiratory analyses were also performed. In addition, the role of non-oxidative glucose pathways (NOGPs), i.e. the polyol pathway, hexosamine biosynthetic pathway (HBP) and PKC were assessed. Key findings These data show that SSB intake: a) resulted in increased weight gain, but did not elicit major effects in terms of insulin resistance and cardiac function after three and six months, respectively; b) triggered myocardial NOGP activation after three months with a reversion after six months; and c) resulted in some impairment in mitochondrial respiratory capacity in response to fatty acid substrate supply after six months. Significance SSB intake did not result in cardiac dysfunction or insulin resistance. However, early changes at the molecular level may increase risk in the longer term.
Collapse
Affiliation(s)
- Natasha Driescher
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Danzil E Joseph
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Veronique R Human
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Edward Ojuka
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Martin Cour
- Hatter Institute for Cardiovascular Research in Africa (HICRA), Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Nkanyiso Hadebe
- Hatter Institute for Cardiovascular Research in Africa (HICRA), Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Dirk Bester
- Oxidative Stress Research Centre, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Jeanine L Marnewick
- Oxidative Stress Research Centre, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa.,Institute of Biomedical and Microbial Biotechnology, Cape Peninsula University of Technology, Bellville, South Africa
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa (HICRA), Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Amanda Lochner
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505, South Africa
| | - M Faadiel Essop
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa
| |
Collapse
|
7
|
Namekawa J, Yasui M, Katayanagi A, Shirai M, Asai F. Increased hepatic triglyceride level induced by a glucokinase activator in mice. ACTA ACUST UNITED AC 2018. [DOI: 10.2131/fts.5.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Junichi Namekawa
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University
- Teijin Pharma Limited
| | | | | | - Mitsuyuki Shirai
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University
| | - Fumitoshi Asai
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University
| |
Collapse
|
8
|
Tsumura Y, Tsushima Y, Tamura A, Hasebe M, Kanou M, Kato H, Kobayashi T. TMG-123, a novel glucokinase activator, exerts durable effects on hyperglycemia without increasing triglyceride in diabetic animal models. PLoS One 2017; 12:e0172252. [PMID: 28207836 PMCID: PMC5313197 DOI: 10.1371/journal.pone.0172252] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 02/01/2017] [Indexed: 12/12/2022] Open
Abstract
Glucokinase (GK) plays a critical role for maintaining glucose homeostasis with regulating glucose uptake in liver and insulin secretion in pancreas. GK activators have been reported to decrease blood glucose levels in patients with type 2 diabetes mellitus. However, clinical development of GK activators has failed due to the loss of glucose-lowering effects and increased plasma triglyceride levels after chronic treatment. Here, we generated a novel GK activator, TMG-123, examined its in vitro and in vivo pharmacological characteristics, and evaluated its risks of aforementioned clinical issues. TMG-123 selectively activated GK enzyme activity without increasing Vmax. TMG-123 improved glucose tolerance without increasing plasma insulin levels in both insulin-deficient (Goto-Kakizaki rats) and insulin-resistant (db/db mice) models. The beneficial effect on glucose tolerance was greater than results observed with clinically available antidiabetic drugs such as metformin and glibenclamide in Zucker Diabetic Fatty rats. TMG-123 also improved glucose tolerance in combination with metformin. After 4 weeks of administration, TMG-123 reduced the Hemoglobin A1c levels without affecting liver and plasma triglyceride levels in Goto-Kakizaki rats and Diet-Induced Obesity mice. Moreover, TMG-123 sustained its effect on Hemoglobin A1c levels even after 24 weeks of administration without affecting triglycerides. Taken together, these data indicate that TMG-123 exerts glucose-lowering effects in both insulin-deficient and -resistant diabetes, and sustains reduced Hemoglobin A1c levels without affecting hepatic and plasma triglycerides even after chronic treatment. Therefore, TMG-123 is expected to be an antidiabetic drug that overcomes the concerns previously reported with other GK activators.
Collapse
Affiliation(s)
- Yoshinori Tsumura
- Pharmaceutical Development Research Laboratories, Teijin Pharma Limited, Hino, Tokyo, Japan
| | - Yu Tsushima
- Pharmaceutical Development Research Laboratories, Teijin Pharma Limited, Hino, Tokyo, Japan
- * E-mail:
| | - Azusa Tamura
- Pharmaceutical Development Research Laboratories, Teijin Pharma Limited, Hino, Tokyo, Japan
| | - Makiko Hasebe
- Pharmaceutical Development Research Laboratories, Teijin Pharma Limited, Hino, Tokyo, Japan
| | - Masanobu Kanou
- Pharmaceutical Development Research Laboratories, Teijin Pharma Limited, Hino, Tokyo, Japan
| | - Hirotsugu Kato
- Pharmaceutical Development Research Laboratories, Teijin Pharma Limited, Hino, Tokyo, Japan
| | - Tsunefumi Kobayashi
- Pharmaceutical Development Research Laboratories, Teijin Pharma Limited, Hino, Tokyo, Japan
| |
Collapse
|
9
|
Wen F, Yang Y, Sun C, Fang H, Nie L, Li L, Liu Y, Yang Z. RESISTIN INHIBITS GLUCOSE-STIMULATED INSULIN SECRETION THROUGH MIR-494 BY TARGET ON STXBP5. ACTA ENDOCRINOLOGICA-BUCHAREST 2017; 13:32-39. [PMID: 31149145 DOI: 10.4183/aeb.2017.32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Aims Resistin has been reported to impair the pancreatic beta cells and associated with insulin resistance. MicroRNAs (miRNAs) are short, endogenously produced non-coding ribonucleotides that bind mRNAs and function mainly as negative regulators in mammals. MiRNAs have been implicated in many diseases, including insulin resistance and diabetes. A considerable body of evidence has indicated an important function for miRNAs in insulin secretion. The current study was designed to investigate the effects of miR-494 in the reductions in insulin secretion attributable to resistin. Methods Insulin secretion was determined by ELISA, and expressions of genes were identified using quantitative RT-PCR (qRT-PCR) or Western blot analysis. Results Insulin secretion was significantly reduced by resistin. Overexpression of miR-494 inhibited insulin secretion both in diet culture and high glucose medium in MIN6 cell lines. MiR-494 down-regulated the protein level of STXBP5 by pairing with sites in the 3'UTR. Conclusion miR-494 is involved in the insulin secretion regulated by resistin via its effects on STXBP5 in MIN6 cells.
Collapse
Affiliation(s)
- F Wen
- Henan University of Science and Technology, College of Animal Science and Technology, Luoyang, Henan, PR China.,Huazhong Agricultural University, College of Life Science and Technology, Breeding and Reproduction of Ministry of Education, Key Laboratory of Agricultural Animal Genetics, Wuhan, Hubei, PR China
| | - Y Yang
- Huazhong Agricultural University, College of Life Science and Technology, Breeding and Reproduction of Ministry of Education, Key Laboratory of Agricultural Animal Genetics, Wuhan, Hubei, PR China
| | - C Sun
- Huazhong Agricultural University, College of Life Science and Technology, Breeding and Reproduction of Ministry of Education, Key Laboratory of Agricultural Animal Genetics, Wuhan, Hubei, PR China
| | - H Fang
- Huazhong Agricultural University, College of Life Science and Technology, Breeding and Reproduction of Ministry of Education, Key Laboratory of Agricultural Animal Genetics, Wuhan, Hubei, PR China
| | - L Nie
- Huazhong Agricultural University, College of Life Science and Technology, Breeding and Reproduction of Ministry of Education, Key Laboratory of Agricultural Animal Genetics, Wuhan, Hubei, PR China
| | - L Li
- Henan University of Science and Technology, College of Animal Science and Technology, Luoyang, Henan, PR China
| | - Y Liu
- Henan University of Science and Technology, College of Animal Science and Technology, Luoyang, Henan, PR China
| | - Z Yang
- Huazhong Agricultural University, College of Life Science and Technology, Breeding and Reproduction of Ministry of Education, Key Laboratory of Agricultural Animal Genetics, Wuhan, Hubei, PR China
| |
Collapse
|
10
|
Nakamura A, Terauchi Y. Present status of clinical deployment of glucokinase activators. J Diabetes Investig 2014; 6:124-32. [PMID: 25802718 PMCID: PMC4364845 DOI: 10.1111/jdi.12294] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 12/14/2022] Open
Abstract
Glucokinase is one of four members of the hexokinase family of enzymes. Its expression is limited to the major organs (such as the pancreas, liver, brain and the gastrointestinal tract) that are thought to have an integrated role in glucose sensing. In the liver, phosphorylation of glucose by glucokinase promotes glycogen synthesis, whereas in the β-cells, it results in insulin release. Studies of glucokinase-linked genetically-modified mice and mutations in humans have illustrated the important roles played by glucokinase in whole-body glucose homeostasis, and suggest that the use of pharmacological agents that augment glucokinase activity could represent a viable treatment strategy in patients with type 2 diabetes. Since 2003, many glucokinase activators (GKAs) have been developed, and their ability to lower the blood glucose has been shown in several animal models of type 2 diabetes. Also, we and others have shown in mouse models that GKAs also have the effect of stimulating the proliferation of β-cells. However, the results of recent phase II trials have shown that GKAs lose their efficacy within several months of use, and that their use is associated with a high incidence of hypoglycemia; furthermore, patients treated with GKAs frequently developed dyslipidemia. A better understanding of the role of glucokinase in metabolic effects is required to resolve several issues identified in clinical trials.
Collapse
Affiliation(s)
- Akinobu Nakamura
- Division of Immunology and Metabolism, Hokkaido University Graduate School of Medicine Sapporo, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University Yokohama, Japan
| |
Collapse
|
11
|
Nakamura A, Shimazaki H, Ohyama S, Eiki J, Terauchi Y. Effect of long-term treatment with a small-molecule glucokinase activator on glucose metabolism, lipid profiles and hepatic function. J Diabetes Investig 2014; 2:276-9. [PMID: 24843498 PMCID: PMC4014967 DOI: 10.1111/j.2040-1124.2011.00104.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We investigated the long‐term effect of a glucokinase (GK) activator (GKA) on the changes in hepatic gene expression, glucose metabolism, lipid profiles and hepatic function in wild‐type mice and the haploinsufficiency of β‐cell‐specific GK mice on a high‐fat (HF) diet. Twenty weeks of GKA treatment had no effect on hepatic GK activity or expression of genes related to glucose or lipid metabolism, suggesting that chronic GK activation by GKA showed a sustained reduction of ambient blood glucose levels without causing significant impact on hepatic lipid and glucose metabolisms. Furthermore, GKA exerted glucose‐lowering efficacy lasted for up to 40 weeks without increasing bodyweight or exerting adverse effects on lipid metabolism or hepatic function in either genotype on the HF diet. The present results show that GKA is capable of chronically improving glucose metabolism in mice on the HF diet without exerting a harmful influence on their lipid profile or hepatic function. (J Diabetes Invest,doi: 10.1111/j.2040‐1124.2011.00103.x, 2011)
Collapse
Affiliation(s)
- Akinobu Nakamura
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama
| | - Hiroko Shimazaki
- Tsukuba Research Institute, Banyu Pharmaceutical Co., Ltd., Tsukuba, Japan
| | - Sumika Ohyama
- Tsukuba Research Institute, Banyu Pharmaceutical Co., Ltd., Tsukuba, Japan
| | - Junichi Eiki
- Tsukuba Research Institute, Banyu Pharmaceutical Co., Ltd., Tsukuba, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama
| |
Collapse
|
12
|
Lu M, Li P, Bandyopadhyay G, Lagakos W, DeWolf WE, Alford T, Chicarelli MJ, Williams L, Anderson DA, Baer BR, McVean M, Conn M, Véniant MM, Coward P. Characterization of a novel glucokinase activator in rat and mouse models. PLoS One 2014; 9:e88431. [PMID: 24533087 PMCID: PMC3922816 DOI: 10.1371/journal.pone.0088431] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 01/07/2014] [Indexed: 12/14/2022] Open
Abstract
Glucokinase (GK) is a hexokinase isozyme that catalyzes the phosphorylation of glucose to glucose-6-phosphate. Glucokinase activators are being investigated as potential diabetes therapies because of their effects on hepatic glucose output and/or insulin secretion. Here, we have examined the efficacy and mechanisms of action of a novel glucokinase activator, GKA23. In vitro, GKA23 increased the affinity of rat and mouse glucokinase for glucose, and increased glucose uptake in primary rat hepatocytes. In vivo, GKA23 treatment improved glucose homeostasis in rats by enhancing beta cell insulin secretion and suppressing hepatic glucose production. Sub-chronic GKA23 treatment of mice fed a high-fat diet resulted in improved glucose homeostasis and lipid profile.
Collapse
Affiliation(s)
- Min Lu
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Pingping Li
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Gautam Bandyopadhyay
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - William Lagakos
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Walter E. DeWolf
- Array BioPharma Inc., Boulder, Colorado, United States of America
| | - Taylor Alford
- Array BioPharma Inc., Boulder, Colorado, United States of America
| | | | - Lance Williams
- Array BioPharma Inc., Boulder, Colorado, United States of America
| | | | - Brian R. Baer
- Array BioPharma Inc., Boulder, Colorado, United States of America
| | - Maralee McVean
- Array BioPharma Inc., Boulder, Colorado, United States of America
| | - Marion Conn
- Amgen Inc., South San Francisco, California, United States of America
| | | | - Peter Coward
- Amgen Inc., South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
13
|
Xiong X, Tao R, DePinho RA, Dong XC. Deletion of hepatic FoxO1/3/4 genes in mice significantly impacts on glucose metabolism through downregulation of gluconeogenesis and upregulation of glycolysis. PLoS One 2013; 8:e74340. [PMID: 24015318 PMCID: PMC3755981 DOI: 10.1371/journal.pone.0074340] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 08/01/2013] [Indexed: 12/16/2022] Open
Abstract
Forkhead transcription factors FoxO1/3/4 have pleiotrophic functions including anti-oxidative stress and metabolism. With regard to glucose metabolism, most studies have been focused on FoxO1. To further investigate their hepatic functions, we generated liver-specific FoxO1/3/4 knockout mice (LTKO) and examined their collective impacts on glucose homeostasis under physiological and pathological conditions. As compared to wild-type mice, LTKO mice had lower blood glucose levels under both fasting and non-fasting conditions and they manifested better glucose and pyruvate tolerance on regular chow diet. After challenged by a high-fat diet, wild-type mice developed type 2 diabetes, but LTKO mice remained euglycemic and insulin-sensitive. To understand the underlying mechanisms, we examined the roles of SIRT6 (Sirtuin 6) and Gck (glucokinase) in the FoxO-mediated glucose metabolism. Interestingly, ectopic expression of SIRT6 in the liver only reduced gluconeogenesis in wild-type but not LTKO mice whereas knockdown of Gck caused glucose intolerance in both wild-type and LTKO mice. The data suggest that both decreased gluconeogenesis and increased glycolysis may contribute to the overall glucose phenotype in the LTKO mice. Collectively, FoxO1/3/4 transcription factors play important roles in hepatic glucose homeostasis.
Collapse
Affiliation(s)
- Xiwen Xiong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Rongya Tao
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Ronald A. DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - X. Charlie Dong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
14
|
Singh H, Giridharan N, Bhonde R, Venkatesan V. Deriving at candidate genes of metabolic stress from pancreas of WNIN/GR-Ob mutant rats. Islets 2013; 5:133-8. [PMID: 24131929 DOI: 10.4161/isl.25520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Development of appropriate animal model systems have greatly helped our understanding of the basic mechanism(s) of several degenerative diseases. WNIN/GR-Ob?a mutant rat strain developed at the National Center for Laboratory Animal Sciences facility of National Institute of Nutrition, is a new animal model ideal to study the metabolic syndrome since it is obese with impaired glucose tolerance and also exhibits several secondary complications. The present study was performed in the pancreas of this mutant model to assess the global gene expression (microarray) to assess the transcriptome level changes in situ depicting inflammation, obesity, insulin resistance, and diabetes in these animals. Our findings suggest an interplay of several confounding factors in pancreas which include inflammation /macrophage infiltration/apoptosis/oxidative and endoplasmic reticulum stress, all contributing for the shift toward pro-inflammation. We were able to phenotypically correlate the metabolic alterations vis-a-vis candidate genes (array analyses) compared between mutants and its age matched, parental controls. We advocate that the data reported here would provide a suitable insight in to the pathophysiology of metabolic syndrome .
Collapse
Affiliation(s)
- Himadri Singh
- Biochemistry/Stem Cell Research; National Institute of Nutrition (ICMR); Tarnaka, Hyderabad, India
| | | | | | | |
Collapse
|
15
|
Kim MY, Jo SH, Park JM, Kim TH, Im SS, Ahn YH. Adenovirus-mediated overexpression of Tcfe3 ameliorates hyperglycaemia in a mouse model of diabetes by upregulating glucokinase in the liver. Diabetologia 2013; 56:635-43. [PMID: 23269357 DOI: 10.1007/s00125-012-2807-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 12/04/2012] [Indexed: 11/30/2022]
Abstract
AIMS/HYPOTHESIS Transcription factor E3 (TFE3) has been shown to increase insulin sensitivity by activating insulin-signalling pathways. However, the role of TFE3 in glucose homeostasis is not fully understood. Here, we explored the possible therapeutic potential of TFE3 for the control of hyperglycaemia using a streptozotocin-induced mouse model of diabetes. METHODS We achieved overabundance of TFE3 in streptozotocin mice by administering an adenovirus (Ad) or adeno-associated virus serotype 2 (AAV2). We also performed an oral glucose tolerance test (OGTT) and insulin tolerance test (ITT). To explore molecular mechanisms of blood glucose control by TFE3, transcriptional studies on the regulation of genes involved in hepatic glucose metabolism were performed using quantitative real-time PCR and chromatin immunoprecipitation assay. The binding site of TFE3 in the liver Gck gene promoter was identified using deletion and site-specific mutation studies. RESULTS Overabundance of TFE3 resulted in reduced hyperglycaemia as shown by the OGTT and ITT in streptozotocin-treated mice. We observed that TFE3 can upregulate Gck in a state of insulin deficiency. However, glucose-6-phosphatase and cytosolic phosphoenolpyruvate carboxykinase mRNA levels were decreased by Ad-mediated overexpression of Tcfe3. Biochemical studies revealed that the anti-hyperglycaemic effect of TFE3 is due to the upregulation of Gck. In primary cultured hepatocytes, TFE3 increased expression of Gck mRNA. Conversely, small interfering RNA-mediated knockdown of TFE3 resulted in a decrease in Gck mRNA. CONCLUSIONS/INTERPRETATION This study demonstrates that TFE3 counteracts hyperglycaemia in streptozotocin-treated mice. This effect could be due to the upregulation of Gck by binding of TFE3 to its cognitive promoter region.
Collapse
Affiliation(s)
- M Y Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-752, Republic of Korea
| | | | | | | | | | | |
Collapse
|
16
|
Winzell MS, Coghlan M, Leighton B, Frangioudakis G, Smith DM, Storlien LH, Ahrén B. Chronic glucokinase activation reduces glycaemia and improves glucose tolerance in high-fat diet fed mice. Eur J Pharmacol 2011; 663:80-6. [DOI: 10.1016/j.ejphar.2011.05.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 04/15/2011] [Accepted: 05/03/2011] [Indexed: 11/29/2022]
|
17
|
Dere E, Lo R, Celius T, Matthews J, Zacharewski TR. Integration of genome-wide computation DRE search, AhR ChIP-chip and gene expression analyses of TCDD-elicited responses in the mouse liver. BMC Genomics 2011; 12:365. [PMID: 21762485 PMCID: PMC3160422 DOI: 10.1186/1471-2164-12-365] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 07/15/2011] [Indexed: 12/20/2022] Open
Abstract
Background The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor (TF) that mediates responses to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Integration of TCDD-induced genome-wide AhR enrichment, differential gene expression and computational dioxin response element (DRE) analyses further elucidate the hepatic AhR regulatory network. Results Global ChIP-chip and gene expression analyses were performed on hepatic tissue from immature ovariectomized mice orally gavaged with 30 μg/kg TCDD. ChIP-chip analysis identified 14,446 and 974 AhR enriched regions (1% false discovery rate) at 2 and 24 hrs, respectively. Enrichment density was greatest in the proximal promoter, and more specifically, within ± 1.5 kb of a transcriptional start site (TSS). AhR enrichment also occurred distal to a TSS (e.g. intergenic DNA and 3' UTR), extending the potential gene expression regulatory roles of the AhR. Although TF binding site analyses identified over-represented DRE sequences within enriched regions, approximately 50% of all AhR enriched regions lacked a DRE core (5'-GCGTG-3'). Microarray analysis identified 1,896 number of TCDD-responsive genes (|fold change| ≥ 1.5, P1(t) > 0.999). Integrating this gene expression data with our ChIP-chip and DRE analyses only identified 625 differentially expressed genes that involved an AhR interaction at a DRE. Functional annotation analysis of differentially regulated genes associated with AhR enrichment identified overrepresented processes related to fatty acid and lipid metabolism and transport, and xenobiotic metabolism, which are consistent with TCDD-elicited steatosis in the mouse liver. Conclusions Details of the AhR regulatory network have been expanded to include AhR-DNA interactions within intragenic and intergenic genomic regions. Moreover, the AhR can interact with DNA independent of a DRE core suggesting there are alternative mechanisms of AhR-mediated gene regulation.
Collapse
Affiliation(s)
- Edward Dere
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | | | |
Collapse
|
18
|
HASHIMOTO H. Study on Establishment of Congenic Strains and Screening of Characteristics in IRS-2 Deficient Mice to Support Translational Research on Type 2 Diabetes. Exp Anim 2011; 60:21-32. [DOI: 10.1538/expanim.60.21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
19
|
Nettleton JA, McKeown NM, Kanoni S, Lemaitre RN, Hivert MF, Ngwa J, van Rooij FJA, Sonestedt E, Wojczynski MK, Ye Z, Tanaka T, Garcia M, Anderson JS, Follis JL, Djousse L, Mukamal K, Papoutsakis C, Mozaffarian D, Zillikens MC, Bandinelli S, Bennett AJ, Borecki IB, Feitosa MF, Ferrucci L, Forouhi NG, Groves CJ, Hallmans G, Harris T, Hofman A, Houston DK, Hu FB, Johansson I, Kritchevsky SB, Langenberg C, Launer L, Liu Y, Loos RJ, Nalls M, Orho-Melander M, Renstrom F, Rice K, Riserus U, Rolandsson O, Rotter JI, Saylor G, Sijbrands EJG, Sjogren P, Smith A, Steingrímsdóttir L, Uitterlinden AG, Wareham NJ, Prokopenko I, Pankow JS, van Duijn CM, Florez JC, Witteman JCM, Dupuis J, Dedoussis GV, Ordovas JM, Ingelsson E, Cupples LA, Siscovick DS, Franks PW, Meigs JB. Interactions of dietary whole-grain intake with fasting glucose- and insulin-related genetic loci in individuals of European descent: a meta-analysis of 14 cohort studies. Diabetes Care 2010; 33:2684-91. [PMID: 20693352 PMCID: PMC2992213 DOI: 10.2337/dc10-1150] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 07/25/2010] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Whole-grain foods are touted for multiple health benefits, including enhancing insulin sensitivity and reducing type 2 diabetes risk. Recent genome-wide association studies (GWAS) have identified several single nucleotide polymorphisms (SNPs) associated with fasting glucose and insulin concentrations in individuals free of diabetes. We tested the hypothesis that whole-grain food intake and genetic variation interact to influence concentrations of fasting glucose and insulin. RESEARCH DESIGN AND METHODS Via meta-analysis of data from 14 cohorts comprising ∼ 48,000 participants of European descent, we studied interactions of whole-grain intake with loci previously associated in GWAS with fasting glucose (16 loci) and/or insulin (2 loci) concentrations. For tests of interaction, we considered a P value <0.0028 (0.05 of 18 tests) as statistically significant. RESULTS Greater whole-grain food intake was associated with lower fasting glucose and insulin concentrations independent of demographics, other dietary and lifestyle factors, and BMI (β [95% CI] per 1-serving-greater whole-grain intake: -0.009 mmol/l glucose [-0.013 to -0.005], P < 0.0001 and -0.011 pmol/l [ln] insulin [-0.015 to -0.007], P = 0.0003). No interactions met our multiple testing-adjusted statistical significance threshold. The strongest SNP interaction with whole-grain intake was rs780094 (GCKR) for fasting insulin (P = 0.006), where greater whole-grain intake was associated with a smaller reduction in fasting insulin concentrations in those with the insulin-raising allele. CONCLUSIONS Our results support the favorable association of whole-grain intake with fasting glucose and insulin and suggest a potential interaction between variation in GCKR and whole-grain intake in influencing fasting insulin concentrations.
Collapse
Affiliation(s)
- Jennifer A Nettleton
- Division of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center, Houston, Houston, Texas, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Liu S, Hatano B, Zhao M, Yen CC, Kang K, Reilly SM, Gangl MR, Gorgun C, Balschi JA, Ntambi JM, Lee CH. Role of peroxisome proliferator-activated receptor {delta}/{beta} in hepatic metabolic regulation. J Biol Chem 2010; 286:1237-47. [PMID: 21059653 DOI: 10.1074/jbc.m110.138115] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pharmacological activation of peroxisome proliferator-activated receptor δ/β (PPARδ/β) improves glucose handling and insulin sensitivity. The target tissues of drug actions remain unclear. We demonstrate here that adenovirus-mediated liver-restricted PPARδ activation reduces fasting glucose levels in chow- and high fat-fed mice. This effect is accompanied by hepatic glycogen and lipid deposition as well as up-regulation of glucose utilization and de novo lipogenesis pathways. Promoter analyses indicate that PPARδ regulates hepatic metabolic programs through both direct and indirect transcriptional mechanisms partly mediated by its co-activator, PPARγ co-activator-1β. Assessment of the lipid composition reveals that PPARδ increases the production of monounsaturated fatty acids, which are PPAR activators, and reduces that of saturated FAs. Despite the increased lipid accumulation, adeno-PPARδ-infected livers exhibit less damage and show a reduction in JNK stress signaling, suggesting that PPARδ-regulated lipogenic program may protect against lipotoxicity. The altered substrate utilization by PPARδ also results in a secondary effect on AMP-activated protein kinase activation, which likely contributes to the glucose-lowering activity. Collectively, our data suggest that PPARδ controls hepatic energy substrate homeostasis by coordinated regulation of glucose and fatty acid metabolism, which provide a molecular basis for developing PPARδ agonists to manage hyperglycemia and insulin resistance.
Collapse
Affiliation(s)
- Sihao Liu
- Department of Genetics and Complex Diseases, Division of Biological Sciences, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Hussain K. Mutations in pancreatic ß-cell Glucokinase as a cause of hyperinsulinaemic hypoglycaemia and neonatal diabetes mellitus. Rev Endocr Metab Disord 2010; 11:179-83. [PMID: 20878480 DOI: 10.1007/s11154-010-9147-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Glucokinase is a key enzyme involved in regulating insulin secretion from the pancreatic ß-cell. The unique role of glucokinase in human glucose physiology is illustrated by the fact that genetic mutations in glucokinase can either cause hyperglycaemia or hypoglycaemia. Heterozygous inactivating mutations in glucokinase cause maturity-onset diabetes of the young (MODY), homozygous inactivating in glucokinase mutations result in permanent neonatal diabetes whereas heterozygous activating glucokinase mutations cause hyperinsulinaemic hypoglycaemia.
Collapse
Affiliation(s)
- Khalid Hussain
- Clinical and Molecular Genetics Unit, The Developmental Endocrinology Research Group, Institute of Child Health, Hospital for Children NHS Trust, University College London, Great Ormond Street, London, UK.
| |
Collapse
|
22
|
Hyperinsulinism and diabetes: genetic dissection of beta cell metabolism-excitation coupling in mice. Cell Metab 2009; 10:442-53. [PMID: 19945402 PMCID: PMC3245718 DOI: 10.1016/j.cmet.2009.10.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 10/13/2009] [Accepted: 10/27/2009] [Indexed: 12/24/2022]
Abstract
The role of metabolism-excitation coupling in insulin secretion has long been apparent, but in recent years, in parallel with studies of human hyperinsulinism and diabetes, genetic manipulation of proteins involved in glucose transport, metabolism, and excitability in mice has brought the central importance of this pathway into sharp relief. We focus on these animal studies and how they provide important insights into not only metabolic and electrical regulation of insulin secretion, but also downstream consequences of alterations in this pathway and the etiology and treatment of insulin-secretion diseases in humans.
Collapse
|
23
|
Nakamura A, Terauchi Y, Ohyama S, Kubota J, Shimazaki H, Nambu T, Takamoto I, Kubota N, Eiki J, Yoshioka N, Kadowaki T, Koike T. Impact of small-molecule glucokinase activator on glucose metabolism and beta-cell mass. Endocrinology 2009; 150:1147-54. [PMID: 19008318 DOI: 10.1210/en.2008-1183] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We investigated the effect of glucokinase activator (GKA) on glucose metabolism and beta-cell mass. We analyzed four mouse groups: wild-type mice and beta-cell-specific haploinsufficiency of glucokinase gene (Gck(+/-)) mice on a high-fat (HF) diet. Each genotype was also treated with GKA mixed in the HF diet. Rodent insulinoma cells and isolated islets were used to evaluate beta-cell proliferation by GKA. After 20 wk on the above diets, there were no differences in body weight, lipid profiles, and liver triglyceride content among the four groups. Glucose tolerance was improved shortly after the GKA treatment in both genotypes of mice. beta-Cell mass increased in wild-type mice compared with Gck(+/-) mice, but a further increase was not observed after the administration of GKA in both genotypes. Interestingly, GKA was able to up-regulate insulin receptor substrate-2 (Irs-2) expression in insulinoma cells and isolated islets. The administration of GKA increased 5-bromo-2-deoxyuridine (BrdU) incorporation in insulinoma cells, and 3 d administration of GKA markedly increased BrdU incorporation in mice treated with GKA in both genotypes, compared with those without GKA. In conclusion, GKA was able to chronically improve glucose metabolism for mice on the HF diet. Although chronic GKA administration failed to cause a further increase in beta-cell mass in vivo, GKA was able to increase beta cell proliferation in vitro and with a 3-d administration in vivo. This apparent discrepancy can be explained by a chronic reduction in ambient blood glucose levels by GKA treatment.
Collapse
Affiliation(s)
- Akinobu Nakamura
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zheng D, Ionut V, Mooradian V, Stefanovski D, Bergman RN. Exenatide sensitizes insulin-mediated whole-body glucose disposal and promotes uptake of exogenous glucose by the liver. Diabetes 2009; 58:352-9. [PMID: 19011168 PMCID: PMC2628608 DOI: 10.2337/db08-0875] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Recent progress suggests that exenatide, a mimetic of glucagon-like peptide-1 (GLP-1), might lower glycemia independent of increased beta-cell response or reduced gastrointestinal motility. We aimed to investigate whether exenatide stimulates glucose turnover directly in insulin-responsive tissues dependent or independent of insulinemia. RESEARCH DESIGN AND METHODS An intraportal glucose infusion clamp was used in dogs to measure glucose turnover to encompass potent activation of the putative glucose/GLP-1 sensor in the porto-hepatic circulation with exenatide. The modified glucose clamp was performed in the presence of postprandial hyperinsulinemia and hyperglycemia with exenatide (20 microg) or saline injected at 0 min. Furthermore, the role of hyperglycemia versus hyperinsulinemia in exenatide-mediated glucose disposal was studied. RESULTS With hyperinsulinemia and hyperglycemia, exenatide produced a significant increase in total glucose turnover by approximately 30%, as indicated by portal glucose infusion rate (saline 15.9 +/- 1.6 vs. exenatide 20.4 +/- 2.1 mg x kg(-1) x min(-1), P < 0.001), resulting from increased whole-body glucose disposal (R(d), approximately 20%) and increased net hepatic uptake of exogenous glucose ( approximately 80%). Reducing systemic hyperglycemia to euglycemia, exenatide still increased total glucose turnover by approximately 20% (saline 13.2 +/- 1.9 vs. exenatide 15.6 +/- 2.1 mg x kg(-1) x min(-1), P < 0.05) in the presence of hyperinsulinemia, accompanied by smaller increments in R(d) (12%) and net hepatic uptake of exogenous glucose (45%). In contrast, reducing hyperinsulinemia to basal levels, exenatide-increased total glucose turnover was completely abolished despite hyperglycemia (saline 2.9 +/- 0.6 vs. exenatide 2.3 +/- 0.3 mg x kg(-1) x min(-1), P = 0.29). CONCLUSIONS Exenatide directly stimulates glucose turnover by enhancing insulin-mediated whole-body glucose disposal and increasing hepatic uptake of exogenous glucose, contributing to its overall action to lower postprandial glucose excursions.
Collapse
Affiliation(s)
- Dan Zheng
- Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | | | | | | | | |
Collapse
|
25
|
Torres TP, Catlin RL, Chan R, Fujimoto Y, Sasaki N, Printz RL, Newgard CB, Shiota M. Restoration of hepatic glucokinase expression corrects hepatic glucose flux and normalizes plasma glucose in zucker diabetic fatty rats. Diabetes 2009; 58:78-86. [PMID: 18952838 PMCID: PMC2606896 DOI: 10.2337/db08-1119] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Accepted: 10/11/2008] [Indexed: 11/13/2022]
Abstract
OBJECTIVE We examined in 20-week-old Zucker diabetic fatty (ZDF) rats whether restoration of hepatic glucokinase (GK) expression would alter hepatic glucose flux and improve hyperglycemia. RESEARCH DESIGN AND METHODS ZDF rats were treated at various doses with an adenovirus that directs the expression of rat liver GK (AdvCMV-GKL) dose dependently, and various metabolic parameters were compared with those of nondiabetic lean littermates (ZCL rats) before and during a hyperglycemic clamp. Viral infection per se did not affect hepatic GK activity, since expression of a catalytically inactive form of GK did not alter endogenous hepatic GK activity. RESULTS ZDF rats compared with ZCL rats have lower hepatic GK activity (11.6 +/- 1.9 vs. 32.5 +/- 3.2 mU/mg protein), marked hyperglycemia (23.9 +/- 1.2 vs. 7.4 +/- 0.3 mmol/l), higher endogenous glucose production (80 +/- 3 vs. 38 +/- 3 micromol x kg(-1) x min(-1)), increased glucose-6-phosphatase flux (150 +/- 11 vs. 58 +/- 8 micromol x kg(-1) x min(-1)), and during a hyperglycemic clamp, a failure to suppress endogenous glucose production (80 +/- 7 vs. -7 +/- 4 micromol x kg(-1) x min(-1)) and promote glucose incorporation into glycogen (15 +/- 5 vs. 43 +/- 3 micromol/g liver). Treatment of ZDF rats with different doses of AdvCMV-GKL, which restored hepatic GK activity to one to two times that of ZCL rats, normalized plasma glucose levels and endogenous glucose production. During a hyperglycemic clamp, glucose production was suppressed and glucose incorporation into glycogen was normal. CONCLUSIONS Alteration of hepatic GK activity in ZDF rats has profound effects on plasma glucose and hepatic glucose flux.
Collapse
Affiliation(s)
- Tracy P Torres
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Gorman T, Hope DCD, Brownlie R, Yu A, Gill D, Löfvenmark J, Wedin M, Mayers RM, Snaith MR, Smith DM. Effect of high-fat diet on glucose homeostasis and gene expression in glucokinase knockout mice. Diabetes Obes Metab 2008; 10:885-97. [PMID: 18034843 DOI: 10.1111/j.1463-1326.2007.00819.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIM We have generated a heterozygous glucokinase knockout mouse (gk(del/wt)), upon which we investigated the effect of high-fat diet (HFD) with respect to metabolic control and both hepatic and beta-cell gene expression. We also investigated the in vitro efficacy of a glucokinase activator (GKA) on glucose-stimulated insulin secretion (GSIS) in gk(del/wt)mouse islets. METHODS Male gk(del/wt)and gk(wt/wt)mice were grouped (n = 8-10) at 10 weeks of age and fed HFD or chow diet (CD) for 10 weeks. Multiple parameters including blood glucose, plasma insulin and glucose tolerance were assessed. Further animal groups were used for in vitro GSIS and islet and liver gene expression analysis. RESULTS AND CONCLUSIONS gk(del/wt)mice showed early-onset persistent hyperglycaemia, raised glycated haemoglobin levels, impaired GSIS and glucose tolerance but no change in plasma cholesterol, non-esterified fatty acids or triglyceride levels. After HFD feeding, insulin levels of gk(del/wt)mice were less than half that of gk(wt/wt)mice, although they were equivalent to gk(wt/wt)mice on CD. While gk(wt/wt)mice maintained moderate hyperglycaemia, gk(del/wt)mice became overtly diabetic, with worsened glucose tolerance. A GKA (GKA50) increased GSIS, at 10 mM glucose, in gk(del/wt)mice to an extent at least as great as that seen in gk(wt/wt)mice on both CD and HFD. gk(del/wt)mice showed only a small number of changes in gene expression compared with gk(wt/wt)mice. We propose the high fat-fed gk(del/wt)mouse as a model of type 2 diabetes and report retained efficacy of a GKA on in vitro GSIS.
Collapse
Affiliation(s)
- T Gorman
- Diabetes & Obesity Drug Discovery, AstraZeneca, Macclesfield, Cheshire, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Type 2 diabetes is a chronic metabolic disease that adversely affects both the quality and longevity of life. The disease is characterised by elevated blood glucose (hyperglycaemia) that is associated with microvascular complications and increased macrovascular risk. Existing oral agents, either alone or in combination, do not exhibit adequate or sustained glucose lowering efficacy in Type 2 diabetics. Consequently, there is an unmet medical need for improved antidiabetic agents which are both more effective at lowering glucose and which display sustained efficacy over a number of years. Such agents would allow present glycaemic treatment targets to be achieved with greater success. Glucokinase activators (GKAs) represent a novel class of glucose-lowering agents. Preclinical data supports the notion that these agents act to lower blood glucose through effects in both the liver and pancreas. It is predicted that this dual compartment mechanism of action of GKAs will translate to robust glucose lowering in diabetic patients. The potential benefits and risks associated with the pharmacological activation of glucokinase are evaluated. The status of GKAs in preclinical and clinical development is assessed are the future prospects of these agents.
Collapse
Affiliation(s)
- Matthew Coghlan
- AstraZeneca Pharmaceuticals, Diabetes and Obesity Drug Discovery, Cardiovascular and Gastrointestinal Research Area, Mereside, Alderley Park, Macclesfi eld SK10 4TG, UK.
| | | |
Collapse
|
28
|
Niu Y, Liu W, Tian C, Xie M, Gao L, Chen Z, Chen X, Li L. Effects of bis(alpha-furancarboxylato)oxovanadium(IV) on glucose metabolism in fat-fed/streptozotocin-diabetic rats. Eur J Pharmacol 2007; 572:213-9. [PMID: 17651728 DOI: 10.1016/j.ejphar.2007.05.071] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Revised: 05/23/2007] [Accepted: 05/24/2007] [Indexed: 11/21/2022]
Abstract
Bis(alpha-furancarboxylato)oxovanadium(IV) (BFOV) is a new orally active anti-diabetic organic vanadium complex. In the previous studies, we found that BFOV exhibited a glucose-lowering activity following oral administration to type 1-like diabetic mice induced by alloxan and rats induced by streptozotocin, and the mechanism was not related to enhancing the insulin synthesis and secretion. Since the enhancement of insulin sensitivity is involved in one of the mechanisms by which vanadium exerts its anti-diabetic effects, BFOV has been further tested on fat-fed/streptozotocin-treated rats, a type 2-like diabetic animal model, in the present study. The results showed that 4 weeks of BFOV treatment significantly improved hyperglycemia, glucose intolerance and hyperinsulinemia, as well as increased insulin sensitivity index in the fat-fed/streptozotocin-diabetic rats. Furthermore, BFOV efficiently activated glucokinase, increased hepatic glycogen content and suppressed phosphoenolpyruvate carboxykinase gene expression in the liver and kidney of the diabetic rats, which contributed to augmentation of hepatic glucose disposal and maintenance of blood glucose homeostasis. These findings suggested that BEOV had anti-diabetic and insulin-sensitizing effects in the diabetic rats, exhibiting the potential to be developed as a new therapeutic agent for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Yanfen Niu
- Yunnan Pharmacological Laboratories of Natural Products, Kunming Medical College, Kunming, PR China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Pino MF, Kim KA, Shelton KD, Lindner J, Odili S, Li C, Collins HW, Shiota M, Matschinsky FM, Magnuson MA. Glucokinase Thermolability and Hepatic Regulatory Protein Binding Are Essential Factors for Predicting the Blood Glucose Phenotype of Missense Mutations. J Biol Chem 2007; 282:13906-16. [PMID: 17353190 DOI: 10.1074/jbc.m610094200] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To better understand how glucokinase (GK) missense mutations associated with human glycemic diseases perturb glucose homeostasis, we generated and characterized mice with either an activating (A456V) or inactivating (K414E) mutation in the gk gene. Animals with these mutations exhibited alterations in their blood glucose concentration that were inversely related to the relative activity index of GK. Moreover, the threshold for glucose-stimulated insulin secretion from islets with either the activating or inactivating mutation were left- or right-shifted, respectively. However, we were surprised to find that mice with the activating mutation had markedly reduced amounts of hepatic GK activity. Further studies of bacterially expressed mutant enzymes revealed that GK(A456V) is as stable as the wild type enzyme, whereas GK(K414E) is thermolabile. However, the ability of GK regulatory protein to inhibit GK(A456V) was found to be less than that of the wild type enzyme, a finding consistent with impaired hepatic nuclear localization. Taken together, this study indicates that it is necessary to have knowledge of both thermolability and the interactions of mutant GK enzymes with GK regulatory protein when attempting to predict in vivo glycemic phenotypes based on the measurement of enzyme kinetics.
Collapse
Affiliation(s)
- Maria F Pino
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Shin JS, Torres TP, Catlin RL, Donahue EP, Shiota M. A defect in glucose-induced dissociation of glucokinase from the regulatory protein in Zucker diabetic fatty rats in the early stage of diabetes. Am J Physiol Regul Integr Comp Physiol 2007; 292:R1381-90. [PMID: 17204595 DOI: 10.1152/ajpregu.00260.2006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Effect of stimulation of glucokinase (GK) export from the nucleus by small amounts of sorbitol on hepatic glucose flux in response to elevated plasma glucose was examined in 6-h fasted Zucker diabetic fatty rats at 10 wk of age. Under basal conditions, plasma glucose, insulin, and glucagon were ∼8 mM, 2,000 pmol/l, and 60 ng/l, respectively. Endogenous glucose production (EGP) was 44 ± 4 μmol·kg−1·min−1. When plasma glucose was raised to ∼17 mM, GK was still predominantly localized with its inhibitory protein in the nucleus. EGP was not suppressed. When sorbitol was infused at 5.6 and 16.7 μmol·kg−1·min−1, along with the increase in plasma glucose, GK was exported to the cytoplasm. EGP (23 ± 19 and 12 ± 5 μmol·kg−1·min−1) was suppressed without a decrease in glucose 6-phosphatase flux (145 ± 23 and 126 ± 16 vs. 122 ± 10 μmol·kg−1·min−1without sorbitol) but increased in glucose phosphorylation as indicated by increases in glucose recycling (122 ± 17 and 114 ± 19 vs. 71 ± 11 μmol·kg−1·min−1), glucose-6-phosphate content (254 ± 32 and 260 ± 35 vs. 188 ± 20 nmol/g liver), fractional contribution of plasma glucose to uridine 5′-diphosphate-glucose flux (43 ± 8 and 42 ± 8 vs. 27 ± 6%), and glycogen synthesis from plasma glucose (20 ± 4 and 22 ± 5 vs. 9 ± 4 μmol glucose/g liver). The decreased glucose effectiveness to suppress EGP and stimulate hepatic glucose uptake may result from failure of the sugar to activate GK by stimulating the translocation of the enzyme.
Collapse
Affiliation(s)
- Jun-Seop Shin
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 702 Light Hall, Nashville, TN 37232-0615, USA
| | | | | | | | | |
Collapse
|
31
|
Efanov AM, Barrett DG, Brenner MB, Briggs SL, Delaunois A, Durbin JD, Giese U, Guo H, Radloff M, Gil GS, Sewing S, Wang Y, Weichert A, Zaliani A, Gromada J. A novel glucokinase activator modulates pancreatic islet and hepatocyte function. Endocrinology 2005; 146:3696-701. [PMID: 15919746 DOI: 10.1210/en.2005-0377] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The glucose-sensing enzyme glucokinase (GK) plays a key role in glucose metabolism. We report here the effects of a novel glucokinase activator, LY2121260. The activator enhanced GK activity via binding to the allosteric site located in the hinge region of the enzyme. LY2121260 stimulated insulin secretion in a glucose-dependent manner in pancreatic beta-cells and increased glucose use in rat hepatocytes. In addition, incubation of beta-cells with the GK activator resulted in increased GK protein levels, suggesting that enhanced insulin secretion on chronic treatment with a GK activator may be due to not only changed enzyme kinetics but also elevated enzyme levels. Animals treated with LY2121260 showed an improved glucose tolerance after oral glucose challenge. These results support the concept that GK activators represent a new class of compounds that increase both insulin secretion and hepatic glucose use and in doing so may prove to be effective agents for the control of blood glucose levels in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Alexander M Efanov
- Lilly Research Laboratories, Eli Lilly & Company, Essener Bogen 7, 22419 Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Postic C, Dentin R, Girard J. Role of the liver in the control of carbohydrate and lipid homeostasis. DIABETES & METABOLISM 2004; 30:398-408. [PMID: 15671906 DOI: 10.1016/s1262-3636(07)70133-7] [Citation(s) in RCA: 300] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The liver plays a unique role in controlling carbohydrate metabolism by maintaining glucose concentrations in a normal range over both short and long periods of times. In type 2 diabetes, alterations in hepatic glucose metabolism are observed, i.e. increased post-absorptive glucose production and impaired suppression of glucose production together with diminished glucose uptake following carbohydrate ingestion. The simultaneous overproduction of glucose and fatty acids in liver further stimulates the secretion of insulin by the pancreatic B cells, and elicits further peripheral insulin resistance thereby establishing a vicious circle. The present review will focus on some of the genetically-altered mouse models that have helped identify enzymes or transcription factors that are essential for maintaining either glucose or lipid homeostasis in liver. Among these mouse models, we will discuss transgenic mice overexpressing key gluconeogenic enzymes (PEPCK, G6Pase) or transcription factors (Foxo1, Pgc1-alpha) that control de novo glucose synthesis. In addition, since the possibility of controlling hepatic glucose utilization as a treatment of type 2 diabetes has been explored we will review some of the strategies proved to be valuable for improving the hyperglycemic phenotype.
Collapse
Affiliation(s)
- C Postic
- Département d'Endocrinologie, Institut Cochin, INSERM U567, CNRS UMR 8104, Université Paris V René Descartes, Paris, France.
| | | | | |
Collapse
|
33
|
Scott DK, Collier JJ, Doan TTT, Bunnell AS, Daniels MC, Eckert DT, O'Doherty RM. A modest glucokinase overexpression in the liver promotes fed expression levels of glycolytic and lipogenic enzyme genes in the fasted state without altering SREBP-1c expression. Mol Cell Biochem 2004; 254:327-37. [PMID: 14674713 DOI: 10.1023/a:1027306122336] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hepatic genes crucial for carbohydrate and lipid homeostasis are regulated by insulin and glucose metabolism. However, the relative contributions of insulin and glucose to the regulation of metabolic gene expression are poorly defined in vivo. To address this issue, adenovirus-mediated hepatic overexpression of glucokinase was used to determine the effects of increased hepatic glucose metabolism on gene expression in fasted or ad libitum fed rats. In the fasted state, a 3 fold glucokinase overexpression was sufficient to mimic feeding-induced increases in pyruvate kinase and acetyl CoA carboxylase mRNA levels, demonstrating a primary role for glucose metabolism in the regulation of these genes in vivo. Conversely, glucokinase overexpression was unable to mimic feeding-induced alterations of fatty acid synthase, glucose-6-phosphate dehydrogenase, carnitine palmitoyl transferase I or PEPCK mRNAs, indicating insulin as the primary regulator of these genes. Interestingly, glucose-6-phosphatase mRNA was increased by glucokinase overexpression in both the fasted and fed states, providing evidence, under these conditions, for the dominance of glucose over insulin signaling for this gene in vivo. Importantly, glucokinase overexpression did not alter sterol regulatory element binding protein 1-c mRNA levels in vivo and glucose signaling did not alter the expression of this gene in primary hepatocytes. We conclude that a modest hepatic overexpression of glucokinase is sufficient to alter expression of metabolic genes without changing the expression of SREBP-1c.
Collapse
Affiliation(s)
- D K Scott
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Gene therapy has been hyped as a possible 'cure' for diabetes mellitus in the near future ever since insulin was first cloned and expressed in cultured cells in the late 1970s. In the past decade, however, the bar for gene therapy for diabetes has been raised because of recent advances in the clinical management of diabetes. Although current treatment modalities fall far short of a cure, they produce greatly improved, if imperfect, glycemic control. In this context, we review the latest advances in in vivo gene therapy and conclude that the most widely applied strategy of insulin gene transfer does not measure up to the existing treatment options, whereas the recently proved concept of induced islet neogenesis has the potential of bettering the currently available therapy. Much work remains to be done, however, before this regimen can be taken from the bench to the bedside.
Collapse
Affiliation(s)
- Lawrence Chan
- Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Texas Medical Center, Houston, TX 77030, USA.
| | | | | |
Collapse
|
35
|
Postic C, Mauvais-Jarvis F, Girard J. Mouse models of insulin resistance and type 2 diabetes. ANNALES D'ENDOCRINOLOGIE 2004; 65:51-9. [PMID: 15122092 DOI: 10.1016/s0003-4266(04)95630-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- C Postic
- Département d'Endocrinologie, Institut Cochin, INSERM U567, CNRS UMR8104 Université Paris V René Descartes, 24, rue du Faubourg Saint Jacques, 75014 Paris, France.
| | | | | |
Collapse
|
36
|
Abstract
Hepatic lipid accumulation may be a result of one or several of the following factors: increased delivery of adipose tissue or dietary fatty acids to the liver, increased de novo synthesis of fatty acids in the liver, decreased rate of hepatic fatty-acid oxidation, or decreased rate in the exit of fatty acids from the liver in the form of triglycerides. Delivery of fatty acids to the liver appears to be the most potent mechanism for hepatic lipid accumulation. Hepatic lipid accumulation is linked to the development of hepatic insulin resistance, which is demonstrated by the impaired suppression of hepatic glucose output by insulin. Current evidence suggests that defects associated with the molecular mechanisms responsible for the propagation of the insulin signal in the liver cells are responsible for the impaired insulin effect and that these defects can develop secondary to lipid accumulation in the liver. Hepatic lipid accumulation appears to affect the activity of phosphatidylinositol 3-kinase, which has a central role in mediating the insulin action in hepatocytes. Generally, exercise has been shown to enhance the insulin action in the liver. Although an exercise-related mechanistic link between attenuation in hepatic lipid accumulation and enhancement in insulin action in the liver has not been described yet, the benefits of exercise on hepatic insulin action may relate to the potential effects of exercise on regulating/preventing hepatic lipid accumulation. However, direct effects of exercise on insulin action in the liver, independent of any effects on hepatic lipid metabolism, cannot currently be excluded. Further research is needed to evaluate the relative importance of exercise in the treatment of hepatic insulin resistance, specifically as it relates to lipid accumulation in the liver.
Collapse
Affiliation(s)
- Christos S Katsanos
- Department of Surgery, The University of Texas Medical Branch, Galveston, Texas, USA.
| |
Collapse
|
37
|
Ferre T, Riu E, Franckhauser S, Agudo J, Bosch F. Long-term overexpression of glucokinase in the liver of transgenic mice leads to insulin resistance. Diabetologia 2003; 46:1662-8. [PMID: 14614559 DOI: 10.1007/s00125-003-1244-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2003] [Revised: 07/28/2003] [Indexed: 10/26/2022]
Abstract
AIMS/HYPOTHESIS Glucokinase overexpression in the liver increases glucose uptake and utilization, and improves glucose tolerance in young transgenic mice. Here, we examined the long-term effects of hepatic overexpression of glucokinase on glucose homeostasis. Moreover, we determined whether glucokinase overexpression counteracted high-fat diet-induced insulin resistance. METHODS Transgenic mice overexpressing glucokinase in liver under the control of the phosphoenolpyruvate carboxykinase promoter, fed either a standard diet or a high-fat diet, were studied. We used non-transgenic littermates as controls. RESULTS Transgenic mice over 6 months old developed impaired glucose tolerance. In addition, at 12 months of age, transgenic mice showed mild hyperglycaemia, hyperinsulinaemia and hypertriglyceridaemia. In spite of increased glucokinase activity, the liver of these mice accumulated less glycogen and increased triglyceride deposition. When 2-month-old glucose-tolerant mice were fed a high-fat diet, transgenic mice gained more body weight and became hyperglycaemic and hyperinsulinaemic. This was concomitant to glucose intolerance, liver steatosis and whole-body insulin resistance. CONCLUSION/INTERPRETATION Long-term overexpression of glucokinase increases hepatic lipogenesis and circulating lipids, which lead to insulin resistance. Our results also suggest that the liver plays a key role in the onset of diabetes.
Collapse
Affiliation(s)
- T Ferre
- Department of Biochemistry and Molecular Biology, School of Veterinary Medicine and Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | | | | | | | | |
Collapse
|
38
|
Otaegui PJ, Ferre T, Riu E, Bosch F. Prevention of obesity and insulin resistance by glucokinase expression in skeletal muscle of transgenic mice. FASEB J 2003; 17:2097-9. [PMID: 14500548 DOI: 10.1096/fj.03-0081fje] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In type 2 diabetes, glucose phosphorylation, a regulatory step in glucose utilization by skeletal muscle, is impaired. Since glucokinase expression in skeletal muscle of transgenic mice increases glucose phosphorylation, we examined whether such mice counteract the obesity and insulin resistance induced by 12 wk of a high-fat diet. When fed this diet, control mice became obese, whereas transgenic mice remained lean. Furthermore, high-fat fed control mice developed hyperglycemia and hyperinsulinemia (a 3-fold increase), indicating that they were insulin resistant. In contrast, transgenic mice were normoglycemic and showed only a mild increase in insulinemia (1.5-fold). They also showed improved whole body glucose tolerance and insulin sensitivity and increased intramuscular concentrations of glucose 6-phosphate and glycogen. A parallel increase in uncoupling protein 3 mRNA levels in skeletal muscle of glucokinase-expressing transgenic mice was also observed. These results suggest that the rise in glucose phosphorylation by glucokinase expression in skeletal muscle leads to increased glucose utilization and energy expenditure that counteracts weight gain and maintains insulin sensitivity.
Collapse
|
39
|
Riu E, Ferre T, Hidalgo A, Mas A, Franckhauser S, Otaegui P, Bosch F. Overexpression of c-myc in the liver prevents obesity and insulin resistance. FASEB J 2003; 17:1715-7. [PMID: 12958186 DOI: 10.1096/fj.02-1163fje] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Alterations in hepatic glucose metabolism play a key role in the development of the hyperglycemia observed in type 2 diabetes. Because the transcription factor c-Myc induces hepatic glucose uptake and utilization and blocks gluconeogenesis, we examined whether hepatic overexpression of c-myc counteracts the insulin resistance induced by a high-fat diet. After 3 months on this diet, control mice became obese, hyperglycemic, and hyperinsulinemic, indicating that they had developed insulin resistance. In contrast, transgenic mice remained lean and showed improved glucose disposal and normal levels of blood glucose and insulin, indicating that they had developed neither obesity nor insulin resistance. These findings were concomitant with normalization of hepatic glucokinase and pyruvate kinase gene expression and enzyme activity, which led to normalization of intrahepatic glucose-6-phosphate and glycogen content. In the liver of control mice fed a high-fat diet, the expression of genes encoding proteins that control energy metabolism, such as sterol receptor element binding protein 1-c, peroxisome proliferator activated receptor alpha, and uncoupling protein-2, was altered. In contrast, in the liver of transgenic mice fed a high-fat diet, the expression of these genes was normal. These results suggest that c-myc overexpression counteracted the obesity and insulin resistance induced by a high-fat diet by modulating the expression of genes that regulate hepatic metabolism.
Collapse
Affiliation(s)
- Efren Riu
- Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, and Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, E-08193-Bellaterra, Spain
| | | | | | | | | | | | | |
Collapse
|
40
|
Affiliation(s)
- Mark A Magnuson
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37205, USA.
| | | | | |
Collapse
|
41
|
Grimsby J, Sarabu R, Corbett WL, Haynes NE, Bizzarro FT, Coffey JW, Guertin KR, Hilliard DW, Kester RF, Mahaney PE, Marcus L, Qi L, Spence CL, Tengi J, Magnuson MA, Chu CA, Dvorozniak MT, Matschinsky FM, Grippo JF. Allosteric activators of glucokinase: potential role in diabetes therapy. Science 2003; 301:370-3. [PMID: 12869762 DOI: 10.1126/science.1084073] [Citation(s) in RCA: 395] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Glucokinase (GK) plays a key role in whole-body glucose homeostasis by catalyzing the phosphorylation of glucose in cells that express this enzyme, such as pancreatic beta cells and hepatocytes. We describe a class of antidiabetic agents that act as nonessential, mixed-type GK activators (GKAs) that increase the glucose affinity and maximum velocity (Vmax) of GK. GKAs augment both hepatic glucose metabolism and glucose-induced insulin secretion from isolated rodent pancreatic islets, consistent with the expression and function of GK in both cell types. In several rodent models of type 2 diabetes mellitus, GKAs lowered blood glucose levels, improved the results of glucose tolerance tests, and increased hepatic glucose uptake. These findings may lead to the development of new drug therapies for diabetes.
Collapse
Affiliation(s)
- Joseph Grimsby
- Department of Metabolic Diseases, Hoffmann-La Roche Inc., Nutley, NJ 07110, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Baudry A, Jackerott M, Lamothe B, Kozyrev SV, Leroux L, Durel B, Saint-Just S, Joshi RL. Partial rescue of insulin receptor-deficient mice by transgenic complementation with an activated insulin receptor in the liver. Gene 2002; 299:219-25. [PMID: 12459269 DOI: 10.1016/s0378-1119(02)01075-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Insulin receptor (IR)-deficient mice develop severe diabetes mellitus, diabetic ketoacidosis (DKA) and liver steatosis and die within 1 week after birth. We examined in this work whether the metabolic phenotype of IR(-/-) mutants could be improved by transgenic complementation with IR selectively in the liver. We first generated transgenic mice expressing a human DNA complementary to RNA encoding a truncated constitutively activated form of IR (IRdelta) under the control of liver-specific phenylalanine hydroxylase (PAH) gene promoter. These mice presented more pronounced fasting hypoglycemia and showed slightly improved glucose tolerance as compared to controls. The transgenic mice were crossed with IR(+/-) mutants to generate IR(-/-) mice carrying the PAH-IRDelta transgene. Although such mutants developed glycosuria, DKA was delayed by more than 1 week and survival was prolonged to 8-20 days in approximately 10% of mice. In these partially rescued pups, serum glucose and triglyceride levels were lowered, hepatic glycogen stores were reconstituted and liver steatosis was absent as compared with pups which developed strong DKA and died earlier. Thus, lack of insulin action in the liver is responsible in large part for the metabolic disorders seen in IR(+/-) mice. This study should stimulate interest in therapeutic strategies aimed at improving hepatic function in diabetes.
Collapse
Affiliation(s)
- Anne Baudry
- Department of Genetics, Development and Molecular Pathology, Institut Cochin, INSERM, CNRS, Université René Decartes, CHU Cochin-Port Royal, 24 Rue du Faubourg Saint-Jacques, 75014 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Morral N, McEvoy R, Dong H, Meseck M, Altomonte J, Thung S, Woo SLC. Adenovirus-mediated expression of glucokinase in the liver as an adjuvant treatment for type 1 diabetes. Hum Gene Ther 2002; 13:1561-70. [PMID: 12228011 DOI: 10.1089/10430340260201653] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Glucokinase (GK) plays a crucial role in hepatic glucose disposal. Its activity is decreased in patients with maturity-onset diabetes of the young and in some animal models of diabetes. We investigated the feasibility of manipulating GK expression as an adjuvant treatment for type 1 diabetes, using an E1/E3-deleted adenoviral vector (Ad.EF1(alpha)GK) delivered to the liver of streptozotocin-induced type 1 diabetic rats. First, we studied the metabolic impact of constitutive glucokinase expression in the absence of insulin. Normal blood glucose levels were observed after gene transfer, and glucose tolerance was substantially enhanced compared with diabetic control animals, suggesting that hepatic GK expression is a feasible mechanism to enhance glucose disposal. In a second study we administered Ad.EF1(alpha)GK together with subcutaneous insulin injections to determine whether the combined action of insulin plus GK activity would provide better glucose homeostasis than insulin treatment alone. This combination approach resulted in constant, near-normal glucose values under fed conditions. Furthermore, the animals stayed in the normoglycemic range after an overnight fast, indicating that the risk to develop hypoglycemia is not increased by expression of GK. Alterations of other metabolic routes were observed, suggesting that insulin-regulated expression of GK may be necessary to use the strategy as a treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Núria Morral
- Carl C. Icahn Institute for Gene Therapy and Molecular Medicine, Mount Sinai School of Medicine, New York, NY 10029-6574, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Guerre-Millo M, Rouault C, Poulain P, André J, Poitout V, Peters JM, Gonzalez FJ, Fruchart JC, Reach G, Staels B. PPAR-alpha-null mice are protected from high-fat diet-induced insulin resistance. Diabetes 2001; 50:2809-14. [PMID: 11723064 DOI: 10.2337/diabetes.50.12.2809] [Citation(s) in RCA: 181] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR)-alpha controls the expression of genes involved in lipid metabolism. PPAR-alpha furthermore participates to maintain blood glucose during acute metabolic stress, as shown in PPAR-alpha-null mice, which develop severe hypoglycemia when fasted. Here, we assessed a potential role for PPAR-alpha in glucose homeostasis in response to long-term high-fat feeding. When subjected to this nutritional challenge, PPAR-alpha-null mice remained normoglycemic and normoinsulinemic, whereas wild-type mice became hyperinsulinemic (190%; P < 0.05) and slightly hyperglycemic (120%; NS). Insulin tolerance tests (ITTs) and glucose tolerance tests (GTTs) were performed to evaluate insulin resistance (IR). Under standard diet, the response to both tests was similar in wild-type and PPAR-alpha-null mice. Under high-fat diet, however, the efficiency of insulin in ITT was reduced and the amount of hyperglycemia in GTT was increased only in wild-type and not in PPAR-alpha-null mice. The IR index, calculated as the product of the areas under glucose and insulin curves in GTT, increased fourfold in high-fat-fed wild-type mice, whereas it remained unchanged in PPAR-alpha-null mice. In contrast, PPAR-alpha deficiency allowed the twofold rise in adiposity and blood leptin levels elicited by the diet. Thus, the absence of PPAR-alpha dissociates IR from high-fat diet-induced increase in adiposity. The effects of PPAR-alpha deficiency on glucose homeostasis seem not to occur via the pancreas, because glucose-stimulated insulin secretion of islets was not influenced by the PPAR-alpha genotype. These data suggest that PPAR-alpha plays a role for the development of IR in response to a Western-type high-fat diet.
Collapse
Affiliation(s)
- M Guerre-Millo
- Unit 465, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Desai UJ, Slosberg ED, Boettcher BR, Caplan SL, Fanelli B, Stephan Z, Gunther VJ, Kaleko M, Connelly S. Phenotypic correction of diabetic mice by adenovirus-mediated glucokinase expression. Diabetes 2001; 50:2287-95. [PMID: 11574410 DOI: 10.2337/diabetes.50.10.2287] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Hyperglycemia of diabetes is caused in part by perturbation of hepatic glucose metabolism. Hepatic glucokinase (GK) is an important regulator of glucose storage and disposal in the liver. GK levels are lowered in patients with maturity-onset diabetes of the young and in some diabetic animal models. Here, we explored the adenoviral vector-mediated overexpression of GK in a diet-induced murine model of type 2 diabetes as a treatment for diabetes. Diabetic mice were treated by intravenous administration with an E1/E2a/E3-deleted adenoviral vector encoding human hepatic GK (Av3hGK). Two weeks posttreatment, the Av3hGK-treated diabetic mice displayed normalized fasting blood glucose levels (95 +/- 4.8 mg/dl; P < 0.001) when compared with Av3Null (135 +/- 5.9 mg/dl), an analogous vector lacking a transgene, and vehicle-treated diabetic mice (134 +/- 8 mg/dl). GK treatment also resulted in lowered insulin levels (632 +/- 399 pg/ml; P < 0.01) compared with the control groups (Av3Null, 1,803 +/- 291 pg/ml; vehicle, 1,861 +/- 392 pg/ml), and the glucose tolerance of the Av3hGK-treated diabetic mice was normalized. No significant increase in plasma or hepatic triglycerides, or plasma free fatty acids was observed in the Av3hGK-treated mice. These data suggest that overexpression of GK may have a therapeutic potential for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- U J Desai
- Genetic Therapy, Inc., Gaithersburg, Maryland. Novartis Institute for Biomedical Research, Summit, New Jersey, USA
| | | | | | | | | | | | | | | | | |
Collapse
|