1
|
Alassaf M, Rajan A. Diet-induced glial insulin resistance impairs the clearance of neuronal debris in Drosophila brain. PLoS Biol 2023; 21:e3002359. [PMID: 37934726 PMCID: PMC10629620 DOI: 10.1371/journal.pbio.3002359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 10/03/2023] [Indexed: 11/09/2023] Open
Abstract
Obesity significantly increases the risk of developing neurodegenerative disorders, yet the precise mechanisms underlying this connection remain unclear. Defects in glial phagocytic function are a key feature of neurodegenerative disorders, as delayed clearance of neuronal debris can result in inflammation, neuronal death, and poor nervous system recovery. Mounting evidence indicates that glial function can affect feeding behavior, weight, and systemic metabolism, suggesting that diet may play a role in regulating glial function. While it is appreciated that glial cells are insulin sensitive, whether obesogenic diets can induce glial insulin resistance and thereby impair glial phagocytic function remains unknown. Here, using a Drosophila model, we show that a chronic obesogenic diet induces glial insulin resistance and impairs the clearance of neuronal debris. Specifically, obesogenic diet exposure down-regulates the basal and injury-induced expression of the glia-associated phagocytic receptor, Draper. Constitutive activation of systemic insulin release from Drosophila insulin-producing cells (IPCs) mimics the effect of diet-induced obesity on glial Draper expression. In contrast, genetically attenuating systemic insulin release from the IPCs rescues diet-induced glial insulin resistance and Draper expression. Significantly, we show that genetically stimulating phosphoinositide 3-kinase (Pi3k), a downstream effector of insulin receptor (IR) signaling, rescues high-sugar diet (HSD)-induced glial defects. Hence, we establish that obesogenic diets impair glial phagocytic function and delays the clearance of neuronal debris.
Collapse
Affiliation(s)
- Mroj Alassaf
- Basic Sciences Division, Fred Hutch, Seattle, Washington, United States of America
| | - Akhila Rajan
- Basic Sciences Division, Fred Hutch, Seattle, Washington, United States of America
| |
Collapse
|
2
|
AMPK inhibits liver gluconeogenesis: fact or fiction? Biochem J 2023; 480:105-125. [PMID: 36637190 DOI: 10.1042/bcj20220582] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/14/2023]
Abstract
Is there a role for AMPK in the control of hepatic gluconeogenesis and could targeting AMPK in liver be a viable strategy for treating type 2 diabetes? These are frequently asked questions this review tries to answer. After describing properties of AMPK and different small-molecule AMPK activators, we briefly review the various mechanisms for controlling hepatic glucose production, mainly via gluconeogenesis. The different experimental and genetic models that have been used to draw conclusions about the role of AMPK in the control of liver gluconeogenesis are critically discussed. The effects of several anti-diabetic drugs, particularly metformin, on hepatic gluconeogenesis are also considered. We conclude that the main effect of AMPK activation pertinent to the control of hepatic gluconeogenesis is to antagonize glucagon signalling in the short-term and, in the long-term, to improve insulin sensitivity by reducing hepatic lipid content.
Collapse
|
3
|
Akalestou E, Lopez-Noriega L, Christakis I, Hu M, Miras AD, Leclerc I, Rutter GA. Vertical sleeve gastrectomy normalizes circulating glucocorticoid levels and lowers glucocorticoid action tissue-selectively in mice. Front Endocrinol (Lausanne) 2022; 13:1020576. [PMID: 36246869 PMCID: PMC9556837 DOI: 10.3389/fendo.2022.1020576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives Glucocorticoids produced by the adrenal cortex are essential for the maintenance of metabolic homeostasis. Glucocorticoid activation is catalysed by 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1). Excess glucocorticoids are associated with insulin resistance and hyperglycaemia. A small number of studies have demonstrated effects on glucocorticoid metabolism of bariatric surgery, a group of gastrointestinal procedures known to improve insulin sensitivity and secretion, which were assumed to result from weight loss. In this study, we hypothesize that a reduction in glucocorticoid action following bariatric surgery contributes to the widely observed euglycemic effects of the treatment. Methods Glucose and insulin tolerance tests were performed at ten weeks post operatively and circulating corticosterone was measured. Liver and adipose tissues were harvested from fed mice and 11β-HSD1 levels were measured by quantitative RT-PCR or Western (immuno-) blotting, respectively. 11β-HSD1 null mice (Hsd11b1 -/-) were generated using CRISPR/Cas9 genome editing. Wild type and littermate Hsd11b1 -/- mice underwent Vertical Sleeve Gastrectomy (VSG) or sham surgery. Results Under the conditions used, no differences in weight loss were observed between VSG treated and sham operated mice. However, both lean and obese WT VSG mice displayed significantly improved glucose clearance and insulin sensitivity. Remarkably, VSG restored physiological corticosterone production in HFD mice and reduced 11β-HSD1 expression in liver and adipose tissue post-surgery. Elimination of the 11β-HSD1/Hsd11b1 gene by CRISPR/Cas9 mimicked the effects of VSG on body weight and tolerance to 1g/kg glucose challenge. However, at higher glucose loads, the euglycemic effect of VSG was superior to Hsd11b1 elimination. Conclusions Bariatric surgery improves insulin sensitivity and reduces glucocorticoid activation at the tissular level, under physiological and pathophysiological (obesity) conditions, irrespective of weight loss. These findings point towards a physiologically relevant gut-glucocorticoid axis, and suggest that lowered glucocorticoid exposure may represent an additional contribution to the health benefits of bariatric surgery.
Collapse
Affiliation(s)
- Elina Akalestou
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Livia Lopez-Noriega
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Ioannis Christakis
- Endocrine and General Surgery, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Ming Hu
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Alexander D. Miras
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Isabelle Leclerc
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Centre de Recherches du CHUM, University of Montreal, Montreal, QC, Canada
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Centre de Recherches du CHUM, University of Montreal, Montreal, QC, Canada
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
4
|
Inhibition of basal and glucagon-induced hepatic glucose production by 991 and other pharmacological AMPK activators. Biochem J 2022; 479:1317-1336. [PMID: 35670459 DOI: 10.1042/bcj20220170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/02/2022] [Accepted: 06/07/2022] [Indexed: 12/24/2022]
Abstract
Pharmacological AMPK activation represents an attractive approach for the treatment of type 2 diabetes (T2D). AMPK activation increases skeletal muscle glucose uptake, but there is controversy as to whether AMPK activation also inhibits hepatic glucose production (HGP) and pharmacological AMPK activators can have off-target effects that contribute to their anti-diabetic properties. The main aim was to investigate the effects of 991 and other direct AMPK activators on HGP and determine whether the observed effects were AMPK-dependent. In incubated hepatocytes, 991 substantially decreased gluconeogenesis from lactate, pyruvate and glycerol, but not from other substrates. Hepatocytes from AMPKβ1-/- mice had substantially reduced liver AMPK activity, yet the inhibition of glucose production by 991 persisted. Also, the glucose-lowering effect of 991 was still seen in AMPKβ1-/- mice subjected to an intraperitoneal pyruvate tolerance test. The AMPK-independent mechanism by which 991 treatment decreased gluconeogenesis could be explained by inhibition of mitochondrial pyruvate uptake and inhibition of mitochondrial sn-glycerol-3-phosphate dehydrogenase-2. However, 991 and new-generation direct small-molecule AMPK activators antagonized glucagon-induced gluconeogenesis in an AMPK-dependent manner. Our studies support the notion that direct pharmacological activation of hepatic AMPK as well as inhibition of pyruvate uptake could be an option for the treatment of T2D-linked hyperglycemia.
Collapse
|
5
|
Dai Y, Shen Y, Guo J, Yang H, Chen F, Zhang W, Wu W, Xu X, Li J. Glycolysis and gluconeogenesis are involved of glucose metabolism adaptation during fasting and re-feeding in black carp (Mylopharyngodon piceus). AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2022.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
6
|
Builes-Montaño CE, Lema-Perez L, Garcia-Tirado J, Alvarez H. Main glucose hepatic fluxes in healthy subjects predicted from a phenomenological-based model. Comput Biol Med 2022; 142:105232. [DOI: 10.1016/j.compbiomed.2022.105232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/08/2022] [Accepted: 01/09/2022] [Indexed: 11/28/2022]
|
7
|
Mohamed YA, Hassaneen HM, El-Dessouky MA, Safwat G, Hassan NAM, Amr K. Study of DYRK1B gene expression and its association with metabolic syndrome in a small cohort of Egyptians. Mol Biol Rep 2021; 48:5497-5502. [PMID: 34291393 DOI: 10.1007/s11033-021-06560-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND A cluster of many risk factors for type 2 diabetes and cardiovascular disease is used to describe the metabolic syndrome (MetS). Moreover, genetic differences associated with metabolic syndrome play a key role in its prevalence and side effects. This study aims to investigate the expression of DYRK1B and its association with metabolic syndrome in a small cohort of Egyptian. MATERIALS AND METHODS A total of 100 adult Egyptians (50 with MetS and 50 healthy control subjects) were included to this study. Clinical, biochemical and anthropometric analysis were assessed. Relative gene expressions of DYRK1B were compared between two groups of subjects using real time PCR. RESULTS We observed marked overexpression in DYRK1B (p < 0.05) in MetS subjects when compared with the healthy control subjects. CONCLUSION This is the first study to provide evidence that DYRK1B is highly expressed among the MetS subjects.
Collapse
Affiliation(s)
- Yara Ahmed Mohamed
- Faculty of Biotechnology, October University for Modern Sciences and Arts University (MSA), No. 12567, 54 Anwar El-Sadat street, Al-Haram, Giza, Egypt.
| | - H M Hassaneen
- Faculty of Science, Chemistry Department, Cairo University, Giza, Egypt
| | | | - Gehan Safwat
- Faculty of Biotechnology, October University for Modern Sciences and Arts University (MSA), No. 12567, 54 Anwar El-Sadat street, Al-Haram, Giza, Egypt
| | - Naglaa Abu-Mandil Hassan
- Medical Research Division, Biological Anthropology Department, National Research Centre, Giza, Egypt
| | - Khalda Amr
- Human Genetics and Genome Research Division, Medical Molecular Genetics Department, National Research Centre, Giza, Egypt
| |
Collapse
|
8
|
Joseph A, Parvathy S, Varma KK, Nandakumar A. Four weeks exercise training enhanced the hepatic insulin sensitivity in high fat- and high carbohydrate-diet fed hyperinsulinemic rats. J Diabetes Metab Disord 2020; 19:1583-1592. [PMID: 33520854 DOI: 10.1007/s40200-020-00694-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/09/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Aim Hyperinsulinemia is considered the primary defect underlying the development of type 2 diabetes. The liver is essential for the regular glucose homeostasis. In this study, we examined the effect of physical training on the insulin signaling, oxidative stress enzymes and Glucose-6-phosphatase(G6Pase) activity in the liver of Wistar rats. Methods Adult male Wistar rats were divided into Control diet group(C), High carbohydrate diet(HCD), High fat diet(HFD), HCD and HFD with training(HCD Ex & HFD Ex). HFD Ex and HCD Ex were trained on a small animal treadmill running at 20 m/min for 30 min, 5 days/wk. The present work investigated the effect of training on hepatic insulin receptor(InsR) signaling events, oxidative stress marker expressions and G6Pase activity in hyperinsulinemic rats. Results High carbohydrate and fat feeding led to hyperinsulinemic status with increased hepatic G6Pase activity and impaired phosphorylation of insulin receptor substrate 1(IRS1) and reduced expression of antioxidant enzymes.Training significantly reduced hepatic G6Pase activity, upregulated phosphoinositide 3 kinase(PI3K) docking site phosphorylation and downregulated the negative IRS1 phosphorylations thereby increasing the glucose transporter(GLUT) expressions (aa(P < 0.001) when compared to HFD, b(P < 0.01),bb (P < 0.001 when compared to HCD). Anti oxidant enzymes like CAT, SOD, eNOS expression were increased with reduction in the expression of inflammatory enzymes like TNF-α and COX-2 (*(P < 0.05),**(P < 0.01),***(P < 0.001) when compared to control, †(P < 0.05),††(P < 0.01),†††(P < 0.001) when compared to HFD and HCD). Conclusion Thus, our study shows that four weeks training enhanced the hepatic insulin sensitivity in high fat and high carbohydrate-diet fed hyperinsulinemic rats. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-020-00694-y.
Collapse
Affiliation(s)
- Anu Joseph
- MIMS Research Foundation, Mankavu P.O., Calicut, Kerala 673007 India
| | - S Parvathy
- MIMS Research Foundation, Mankavu P.O., Calicut, Kerala 673007 India
| | | | | |
Collapse
|
9
|
Rasal KD, Iquebal MA, Dixit S, Vasam M, Raza M, Sahoo L, Jaiswal S, Nandi S, Mahapatra KD, Rasal A, Udit UK, Meher PK, Murmu K, Angadi UB, Rai A, Kumar D, Sundaray JK. Revealing Alteration in the Hepatic Glucose Metabolism of Genetically Improved Carp, Jayanti Rohu Labeo rohita Fed a High Carbohydrate Diet Using Transcriptome Sequencing. Int J Mol Sci 2020; 21:E8180. [PMID: 33142948 PMCID: PMC7662834 DOI: 10.3390/ijms21218180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/25/2023] Open
Abstract
Although feed cost is the greatest concern in aquaculture, the inclusion of carbohydrates in the fish diet, and their assimilation, are still not well understood in aquaculture species. We identified molecular events that occur due to the inclusion of high carbohydrate levels in the diets of genetically improved 'Jayanti rohu' Labeo rohita. To reveal transcriptional changes in the liver of rohu, a feeding experiment was conducted with three doses of gelatinized starch (20% (control), 40%, and 60%). Transcriptome sequencing revealed totals of 15,232 (4464 up- and 4343 down-regulated) and 15,360 (4478 up- and 4171 down-regulated) differentially expressed genes. Up-regulated transcripts associated with glucose metabolisms, such as hexokinase, PHK, glycogen synthase and PGK, were found in fish fed diets with high starch levels. Interestingly, a de novo lipogenesis mechanism was found to be enriched in the livers of treated fish due to up-regulated transcripts such as FAS, ACCα, and PPARγ. The insulin signaling pathways with enriched PPAR and mTOR were identified by Kyoto Encyclopedia of Genes and Genome (KEGG) as a result of high carbohydrates. This work revealed for the first time the atypical regulation transcripts associated with glucose metabolism and lipogenesis in the livers of Jayanti rohu due to the inclusion of high carbohydrate levels in the diet. This study also encourages the exploration of early nutritional programming for enhancing glucose efficiency in carp species, for sustainable and cost-effective aquaculture production.
Collapse
Affiliation(s)
- Kiran D. Rasal
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Mir Asif Iquebal
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Sangita Dixit
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Manohar Vasam
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Mustafa Raza
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Lakshman Sahoo
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Sarika Jaiswal
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Samiran Nandi
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Kanta Das Mahapatra
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Avinash Rasal
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Uday Kumar Udit
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Prem Kumar Meher
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Khuntia Murmu
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - UB Angadi
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Anil Rai
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Dinesh Kumar
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Jitendra Kumar Sundaray
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| |
Collapse
|
10
|
Akalestou E, Genser L, Rutter GA. Glucocorticoid Metabolism in Obesity and Following Weight Loss. Front Endocrinol (Lausanne) 2020; 11:59. [PMID: 32153504 PMCID: PMC7045057 DOI: 10.3389/fendo.2020.00059] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/30/2020] [Indexed: 01/08/2023] Open
Abstract
Glucocorticoids are steroid hormones produced by the adrenal cortex and are essential for the maintenance of various metabolic and homeostatic functions. Their function is regulated at the tissue level by 11β-hydroxysteroid dehydrogenases and they signal through the glucocorticoid receptor, a ligand-dependent transcription factor. Clinical observations have linked excess glucocorticoid levels with profound metabolic disturbances of intermediate metabolism resulting in abdominal obesity, insulin resistance and dyslipidaemia. In this review, we discuss the physiological mechanisms of glucocorticoid secretion, regulation and function, and survey the metabolic consequences of excess glucocorticoid action resulting from elevated release and activation or up-regulated signaling. Finally, we summarize the reported impact of weight loss by diet, exercise, or bariatric surgery on circulating and tissue-specific glucocorticoid levels and examine the therapeutic possibility of reversing glucocorticoid-associated metabolic disorders.
Collapse
Affiliation(s)
- Elina Akalestou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
| | - Laurent Genser
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
- Department of Digestive and Hepato-Pancreato-Biliary Surgery, Liver Transplantation, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière University Hospital, Institut Hospitalo-Universitaire ICAN, Sorbonne Université, Paris, France
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
- *Correspondence: Guy A. Rutter
| |
Collapse
|
11
|
El-Sonbaty YA, Suddek GM, Megahed N, Gameil NM. Protocatechuic acid exhibits hepatoprotective, vasculoprotective, antioxidant and insulin-like effects in dexamethasone-induced insulin-resistant rats. Biochimie 2019; 167:119-134. [PMID: 31557503 DOI: 10.1016/j.biochi.2019.09.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 09/16/2019] [Indexed: 12/28/2022]
Abstract
Protocatechuic acid (PCA), the natural phenolic antioxidant, reportedly exhibited hypoglycemic and insulin-like effects. Recent studies have reported its cardioprotective effect in glucocorticoid (GC)-induced hypertensive rats. Nevertheless, its beneficial role has not been investigated in the setting of GCs excess-induced insulin resistance. This study aimed to investigate the possible protective potential and the plausible mechanisms of pretreatment with PCA against GCs-induced insulin resistance, liver steatosis and vascular dysfunction. Insulin resistance was induced in male Wistar rats by a 7-day treatment with dexamethasone (DEX) (1 mg/kg/day, i.p.). PCA (50, 100 mg/kg/day, orally) was started 7 days before DEX administration and continued during the test period. PCA significantly and dose-dependently attenuated DEX-induced a) glucose intolerance (↓ AUCOGTT), b) hyperglycemia (↓ fasting blood glucose), c) impaired insulin sensitivity [↓fasting plasma insulin and homeostasis model assessment of insulin resistance (HOMA-IR) index)] and d) dyslipidemia (↓total cholesterol, triglycerides, low-density lipoprotein-cholesterol and very low-density lipoprotein-cholesterol). PCA mitigated DEX-induced liver steatosis with associated reduction in serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activity. Moreover, PCA ameliorated DEX-induced vascular dysfunction and enhanced ACh-induced relaxation in aortic rings. The metabolic ameliorating effects of PCA might be attributed to the enhanced insulin signaling in soleus muscles (↑AKT phosphorylation) and mitigating gluconeogenesis (↓ hepatic mRNA expression of phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase). The vasculoprotective effect of PCA might be related to its ability to restore normal mRNA expression of [endothelial nitric oxide synthase (eNOS) and NADPH Oxidase 4 (NOX4)]. PCA restored normal oxidative balance [↓ oxidant species, malondialdehyde (MDA) and (↑ antioxidant superoxide dismutase (SOD)]. The findings herein reveal for the first time that PCA may be taken as a supplement with GCs to limit their metabolic and vascular side effects through its hypoglycemic, insulin-sensitizing, hypolipidemic and antioxidant effects.
Collapse
Affiliation(s)
- Yomna A El-Sonbaty
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Ghada M Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Nirmeen Megahed
- Department of Pathology, Faculty of Medicine, Mansoura University, Egypt
| | - Nariman M Gameil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
12
|
Sheikh Y, Chanu MB, Mondal G, Manna P, Chattoraj A, Chandra Deka D, Chandra Talukdar N, Chandra Borah J. Procyanidin A2, an anti-diabetic condensed tannin extracted from Wendlandia glabrata, reduces elevated G-6-Pase and mRNA levels in diabetic mice and increases glucose uptake in CC1 hepatocytes and C1C12 myoblast cells. RSC Adv 2019; 9:17211-17219. [PMID: 35519885 PMCID: PMC9064588 DOI: 10.1039/c9ra02397f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 05/14/2019] [Indexed: 12/26/2022] Open
Abstract
To reduce the global burden of diabetes in an affordable way great attention has been paid to the search for functional foods and herbal remedies. One of the most popularly used functional foods in the North Eastern region of India is tender shoots of Wendlandia glabrata DC. In the current study identification of active anti-diabetic constituent of the tender shoots of W. glabrata was guided through α-glucosidase inhibition and procyanidin A2 was identified with IC50 0.27 ± 0.01 μg mL−1 making it potential source for postprandial management of DM type 2. The study has also demonstrated procyanidin A2 as a potent anti-diabetic agent that exhibits significant glucose-6-phosphatase inhibitory activities and downregulated mRNA level in diabetic mice as well as increases glucose uptake in hepatocytes and myoblast cells. This study revealed that easily available tender shoots of W. glabrata could be used to make specific dietary recommendations for consumption for affordable management of diabetes. Wendlandia glabrata and procyanidin A2 isolated thereof are exhibited significant anti-diabetic effect.![]()
Collapse
Affiliation(s)
- Yunush Sheikh
- Institute of Buioresources and Sustainable Development Imphal-795001 Manipur India
| | - Maibam Beebina Chanu
- Institute of Buioresources and Sustainable Development Imphal-795001 Manipur India
| | - Gopinath Mondal
- Institute of Buioresources and Sustainable Development Imphal-795001 Manipur India
| | - Prasenjit Manna
- CSIR-North East Institute of Science and Technology Jorhat-785006 Assam India
| | - Asamanja Chattoraj
- Institute of Buioresources and Sustainable Development Imphal-795001 Manipur India
| | | | - Narayan Chandra Talukdar
- Laboratory of Natural Medicinal Chemistry, Life Sciences Division, Institute of Advanced Study in Science & Technology Guwahati-781035 Assam India +91-361-2273063 +91-361-2273061
| | - Jagat Chandra Borah
- Laboratory of Natural Medicinal Chemistry, Life Sciences Division, Institute of Advanced Study in Science & Technology Guwahati-781035 Assam India +91-361-2273063 +91-361-2273061
| |
Collapse
|
13
|
Berndt N, Holzhütter HG. Dynamic Metabolic Zonation of the Hepatic Glucose Metabolism Is Accomplished by Sinusoidal Plasma Gradients of Nutrients and Hormones. Front Physiol 2018; 9:1786. [PMID: 30631280 PMCID: PMC6315134 DOI: 10.3389/fphys.2018.01786] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/28/2018] [Indexed: 12/19/2022] Open
Abstract
Being the central metabolic organ of vertebrates, the liver possesses the largest repertoire of metabolic enzymes among all tissues and organs. Almost all metabolic pathways are resident in the parenchymal cell, hepatocyte, but the pathway capacities may largely differ depending on the localization of hepatocytes within the liver acinus-a phenomenon that is commonly referred to as metabolic zonation. Metabolic zonation is rather dynamic since gene expression patterns of metabolic enzymes may change in response to nutrition, drugs, hormones and pathological states of the liver (e.g., fibrosis and inflammation). This fact has to be ultimately taken into account in mathematical models aiming at the prediction of metabolic liver functions in different physiological and pathological settings. Here we present a spatially resolved kinetic tissue model of hepatic glucose metabolism which includes zone-specific temporal changes of enzyme abundances which are driven by concentration gradients of nutrients, hormones and oxygen along the hepatic sinusoids. As key modulators of enzyme expression we included oxygen, glucose and the hormones insulin and glucagon which also control enzyme activities by cAMP-dependent reversible phosphorylation. Starting with an initially non-zonated model using plasma profiles under fed, fasted and diabetic conditions, zonal patterns of glycolytic and gluconeogenetic enzymes as well as glucose uptake and release rates are created as an emergent property. We show that mechanisms controlling the adaptation of enzyme abundances to varying external conditions necessarily lead to the zonation of hepatic carbohydrate metabolism. To the best of our knowledge, this is the first kinetic tissue model which takes into account in a semi-mechanistic way all relevant levels of enzyme regulation.
Collapse
Affiliation(s)
- Nikolaus Berndt
- Computational Biochemistry Group, Institute of Biochemistry, Charite-University Medicine Berlin, Berlin, Germany.,Institute for Computational and Imaging Science in Cardiovascular Medicine, Charite-University Medicine Berlin, Berlin, Germany
| | - Hermann-Georg Holzhütter
- Computational Biochemistry Group, Institute of Biochemistry, Charite-University Medicine Berlin, Berlin, Germany
| |
Collapse
|
14
|
Logie L, Lees Z, Allwood JW, McDougall G, Beall C, Rena G. Regulation of hepatic glucose production and AMPK by AICAR but not by metformin depends on drug uptake through the equilibrative nucleoside transporter 1 (ENT1). Diabetes Obes Metab 2018; 20:2748-2758. [PMID: 29962100 PMCID: PMC6282725 DOI: 10.1111/dom.13455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/28/2018] [Accepted: 06/28/2018] [Indexed: 01/15/2023]
Abstract
AIM Recently we have observed differences in the ability of metformin and AICAR to repress glucose production from hepatocytes using 8CPT-cAMP. Previous results indicate that, in addition to activating protein kinase A, 8CPT-modified cAMP analogues suppress the nitrobenzylthioinosine (NBMPR)-sensitive equilibrative nucleoside transporter ENT1. We aimed to exploit 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR, which is highly selective for a high-affinity binding-site on ENT1, to investigate the role of ENT1 in the liver-specific glucose-lowering properties of AICAR and metformin. METHODS Primary mouse hepatocytes were incubated with AICAR and metformin in combination with cAMP analogues, glucagon, forskolin and NBMPR. Hepatocyte glucose production (HGP) and AMPK signalling were measured, and a uridine uptake assay with supporting LC-MS was used to investigate nucleoside depletion from medium by cells. RESULTS AICAR and metformin increased AMPK pathway phosphorylation and decreased HGP induced by dibutyryl cAMP and glucagon. HGP was also induced by 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR; however, in each case this was resistant to suppression by AICAR but not by metformin. Cross-validation of tracer and mass spectrometry studies indicates that 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR inhibited the effects of AICAR, at least in part, by impeding its uptake into hepatocytes. CONCLUSIONS We report for the first time that suppression of ENT1 induces HGP. ENT1 inhibition also impedes uptake and the effects of AICAR, but not metformin, on HGP. Further investigation of nucleoside transport may illuminate a better understanding of how metformin and AICAR each regulate HGP.
Collapse
Affiliation(s)
- Lisa Logie
- Division of Cellular MedicineNinewells Hospital and Medical School, University of DundeeDundeeUK
| | - Zoe Lees
- Division of Cellular MedicineNinewells Hospital and Medical School, University of DundeeDundeeUK
- Environmental and Biochemical SciencesThe James Hutton InstituteDundeeUK
| | - J. William Allwood
- Environmental and Biochemical SciencesThe James Hutton InstituteDundeeUK
| | - Gordon McDougall
- Environmental and Biochemical SciencesThe James Hutton InstituteDundeeUK
| | - Craig Beall
- Institute of Biomedical and Clinical ScienceUniversity of Exeter Medical SchoolExeterUK
| | - Graham Rena
- Division of Cellular MedicineNinewells Hospital and Medical School, University of DundeeDundeeUK
| |
Collapse
|
15
|
Honma M, Sawada S, Ueno Y, Murakami K, Yamada T, Gao J, Kodama S, Izumi T, Takahashi K, Tsukita S, Uno K, Imai J, Kakazu E, Kondo Y, Mizuno K, Kawagishi N, Shimosegawa T, Katagiri H. Selective insulin resistance with differential expressions of IRS-1 and IRS-2 in human NAFLD livers. Int J Obes (Lond) 2018; 42:1544-1555. [PMID: 29717275 PMCID: PMC6160396 DOI: 10.1038/s41366-018-0062-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 01/31/2018] [Accepted: 02/13/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND/OBJECTIVE Insulin signals, via the regulation of key enzyme expression, both suppress gluconeogenesis and enhance lipid synthesis in the liver. Animal studies have revealed insulin signaling favoring gluconeogenesis suppression to be selectively impaired in steatotic livers. However, whether, and if so how, such selective insulin resistance occurs in human steatotic livers remains unknown. Our aim was to investigate selective insulin resistance in human livers with non-alcoholic fatty liver disease (NAFLD). SUBJECTS/METHODS We examined mRNA expressions of key molecules for insulin signaling, gluconeogenesis and lipogenesis in human liver biopsy samples obtained from 51 non-diabetic subjects: 9 healthy controls and 42 NAFLD patients, and analyzed associations of these molecules with each other and with detailed pathological and clinical biochemistry data. RESULTS In NAFLD patients, insulin receptor substrate (IRS)-2 expression was decreased, while those of key enzymes for gluconeogenesis were increased. These alterations of IRS-2 and gluconeogenesis enzymes were induced both in simple steatosis (SS) and non-alcoholic steatohepatitis (NASH), while these expression levels did not differ between SS and NASH. Furthermore, alterations in the expressions of IRS-2 and gluconeogenesis enzymes showed strong negative correlations and were concurrently induced in the early histological stage of NAFLD. In contrast, fatty acid synthase (FAS) expression was not decreased in NAFLD, despite IRS-2 downregulation, but correlated strongly with IRS-1 expression. Furthermore, no histological scores were associated with these molecules. Thus, IRS-1 signaling, which is not impaired in NAFLD, appears to modulate FAS expression. CONCLUSION These analyses revealed that selective insulin resistance is present in human NAFLD livers and occurs in its early phases. The effect of insulin, during the IRS step, on gene expressions for lipogenesis and gluconeogenesis are apparently distinct and preferential downregulation of IRS-2 may contribute to selective resistance to the suppressive effects of insulin on gluconeogenesis.
Collapse
Affiliation(s)
- Midori Honma
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shojiro Sawada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Yoshiyuki Ueno
- Department of Gastroenterology, Yamagata University Faculty of Medicine, Yamagata, Japan
- Japan Agency for Medical Research and Development, CREST, Tokyo, Japan
| | - Keigo Murakami
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuya Yamada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
- Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junhong Gao
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinjiro Kodama
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohito Izumi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sohei Tsukita
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | - Eiji Kakazu
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuteru Kondo
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Mizuno
- Department of Gastroenterology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Naoki Kawagishi
- Division of Transplantation, Reconstruction and Endoscopic Surgery, Tohoku University Hospital, Sendai, Japan
| | - Tooru Shimosegawa
- Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan.
- Japan Agency for Medical Research and Development, CREST, Tokyo, Japan.
- Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
16
|
Joshi AKR, Kandlakunta B, Kotturu SK, Ghosh S. Antiglucocorticoid potential of nutraceuticals: In silico molecular docking and in vitro assessment. J Food Biochem 2018. [DOI: 10.1111/jfbc.12522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Apurva Kumar Ramesh Joshi
- Food Chemistry Division; National Institute of Nutrition, Jamai-Osmania; Telangana Hyderabad 500007 India
| | - Bhaskarachary Kandlakunta
- Food Chemistry Division; National Institute of Nutrition, Jamai-Osmania; Telangana Hyderabad 500007 India
| | - Sandeep Kumar Kotturu
- Division of Molecular Biology; National Institute of Nutrition, Jamai-Osmania; Telangana Hyderabad 500007 India
| | - Sudip Ghosh
- Division of Molecular Biology; National Institute of Nutrition, Jamai-Osmania; Telangana Hyderabad 500007 India
| |
Collapse
|
17
|
Li J, Yu H, Wang S, Wang W, Chen Q, Ma Y, Zhang Y, Wang T. Natural products, an important resource for discovery of multitarget drugs and functional food for regulation of hepatic glucose metabolism. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:121-135. [PMID: 29391777 PMCID: PMC5768189 DOI: 10.2147/dddt.s151860] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Imbalanced hepatic glucose homeostasis is one of the critical pathologic events in the development of metabolic syndromes (MSs). Therefore, regulation of imbalanced hepatic glucose homeostasis is important in drug development for MS treatment. In this review, we discuss the major targets that regulate hepatic glucose homeostasis in human physiologic and pathophysiologic processes, involving hepatic glucose uptake, glycolysis and glycogen synthesis, and summarize their changes in MSs. Recent literature suggests the necessity of multitarget drugs in the management of MS disorder for regulation of imbalanced glucose homeostasis in both experimental models and MS patients. Here, we highlight the potential bioactive compounds from natural products with medicinal or health care values, and focus on polypharmacologic and multitarget natural products with effects on various signaling pathways in hepatic glucose metabolism. This review shows the advantage and feasibility of discovering multicompound-multitarget drugs from natural products, and providing a new perspective of ways on drug and functional food development for MSs.
Collapse
Affiliation(s)
- Jian Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin
| | - Haiyang Yu
- Department of Phytochemistry, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Sijian Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin
| | - Wei Wang
- Internal Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Qian Chen
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin
| | - Yanmin Ma
- Department of Phytochemistry, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin
| | - Tao Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin
| |
Collapse
|
18
|
Chen YJ, Zhang TY, Chen HY, Lin SM, Luo L, Wang DS. Simultaneous stimulation of glycolysis and gluconeogenesis by feeding in the anterior intestine of the omnivorous GIFT tilapia, Oreochromis niloticus. Biol Open 2017; 6:818-824. [PMID: 28619994 PMCID: PMC5483027 DOI: 10.1242/bio.024836] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The present study was performed to investigate the roles of anterior intestine in the postprandial glucose homeostasis of the omnivorous Genetically Improved Farmed Tilapia (GIFT). Sub-adult fish (about 173 g) were sampled at 0, 1, 3, 8 and 24 h post feeding (HPF) after 36 h of food deprivation, and the time course of changes in intestinal glucose transport, glycolysis, glycogenesis and gluconeogenesis at the transcription and enzyme activity level, as well as plasma glucose contents, were analyzed. Compared with 0 HPF (fasting for 36 h), the mRNA levels of both ATP-dependent sodium/glucose cotransporter 1 and facilitated glucose transporter 2 increased during 1-3 HPF, decreased at 8 HPF and then leveled off. These results indicated that intestinal uptake of glucose and its transport across the intestine to blood mainly occurred during 1-3 HPF, which subsequently resulted in the increase of plasma glucose level at the same time. Intestinal glycolysis was stimulated during 1-3 HPF, while glucose storage as glycogen was induced during 3-8 HPF. Unexpectedly, intestinal gluconeogenesis (IGNG) was also strongly induced during 1-3 HPF at the state of nutrient assimilation. The mRNA abundance and enzyme activities of glutamic-pyruvic and glutamic-oxaloacetic transaminases increased during 1-3 HPF, suggesting that the precursors of IGNG might originate from some amino acids. Taken together, it was concluded that the anterior intestine played an important role in the regulation of postprandial glucose homeostasis in omnivorous tilapia, as it represented significant glycolytic potential and glucose storage. It was interesting that postprandial IGNG was stimulated by feeding temporarily, and its biological significance remains to be elucidated in fish.
Collapse
Affiliation(s)
- Yong-Jun Chen
- Key Laboratory of Freshwater Fish Resources and Reproductive Development (Ministry of Education), College of Animal Science and Technology, Southwest University, Chongqing 400715, China .,Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Ti-Yin Zhang
- Key Laboratory of Freshwater Fish Resources and Reproductive Development (Ministry of Education), College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Hai-Yan Chen
- Key Laboratory of Freshwater Fish Resources and Reproductive Development (Ministry of Education), College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Shi-Mei Lin
- Key Laboratory of Freshwater Fish Resources and Reproductive Development (Ministry of Education), College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Li Luo
- Key Laboratory of Freshwater Fish Resources and Reproductive Development (Ministry of Education), College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - De-Shou Wang
- Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
19
|
3'-UTR SNP rs2229611 in G6PC1 affects mRNA stability, expression and Glycogen Storage Disease type-Ia risk. Clin Chim Acta 2017; 471:46-54. [PMID: 28502559 DOI: 10.1016/j.cca.2017.05.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/09/2017] [Accepted: 05/11/2017] [Indexed: 12/18/2022]
Abstract
The frequency of rs2229611, previously reported in Chinese, Caucasians, Japanese and Hispanics, was investigated for the first time in Indian ethnicity. We analyzed its role in the progression of Glycogen Storage Disease type-Ia (GSD-Ia) and breast cancer. Genotype data on rs2229611 revealed that the risk of GSD-Ia was higher (P=0.0195) with CC compared to TT/TC genotypes, whereas no such correlation was observed with breast cancer cases. We observed a strong linkage disequilibrium (LD) among rs2229611 and other disease causing G6PC1 variants (|D'|=1, r2=1). Functional validation performed in HepG2 cells using luciferase constructs showed significant (P<0.05) decrease in expression than wild-type 3'-UTR due to curtailed mRNA stability. Furthermore, AU-rich elements (AREs) mediated regulation of G6PC1 expression characterized using 3'-UTR deletion constructs showed a prominent decrease in mRNA stability. We then examined whether miRNAs are involved in controlling G6PC1 expression using pmirGLO-UTR constructs, with evidence of more distinct inhibition in the reporter function with rs2229611. These data suggests that rs2229611 is a crucial regulatory SNP which in homozygous state leads to a more aggressive disease phenotype in GSD-Ia patients. The implication of this result is significant in predicting disease onset, progression and response to disease modifying treatments in patients with GSD-Ia.
Collapse
|
20
|
Xiong XP, Song Q, Han CC, Gan W, He F, Wei SH, Liu HH, Xu HY. Insulin Promotes the Expression of the Gluconeogenic Rate-Limiting Enzymes Phosphoenolpyruvate Carboxykinase (Pepck) and Glucose 6-Phosphatase (G6pase) through PI3k/Akt/mTOR Signaling Pathway in Goose Hepatocytes. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2016. [DOI: 10.1590/1806-9061-2015-0080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- XP Xiong
- Sichuan Agricultural University, China
| | - Q Song
- Sichuan Agricultural University, China
| | - CC Han
- Sichuan Agricultural University, China
| | - W Gan
- Sichuan Agricultural University, China
| | - F He
- Sichuan Agricultural University, China
| | - SH Wei
- Sichuan Agricultural University, China
| | - HH Liu
- Sichuan Agricultural University, China
| | - HY Xu
- Sichuan Agricultural University, China
| |
Collapse
|
21
|
Chen G, Wang R, Chen H, Wu L, Ge RS, Wang Y. Gossypol ameliorates liver fibrosis in diabetic rats induced by high-fat diet and streptozocin. Life Sci 2016; 149:58-64. [PMID: 26883980 DOI: 10.1016/j.lfs.2016.02.044] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 02/07/2016] [Accepted: 02/10/2016] [Indexed: 02/06/2023]
Abstract
11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) inhibitors have been shown to treat type 2 diabetes (T2D). Since gossypol is an 11β-HSD1 inhibitor, the objective of the present study was to treat T2D and T2D-related liver fibrosis in rat model using low-dose gossypol. T2D was induced by feeding with high fat diet plus injection of streptozocin (30mg/kg). Diabetic rats were treated with either vehicle control or racemic gossypol with a dose of 15mg/kg/day for 4weeks followed by 15mg/kg/week for additional 8weeks. Blood glucose, cholesterol, LDL, and triglycerides were measured. Messenger mRNA levels of glucocorticoid receptor (Nr3c1), phosphoenolpyruvate carboxykinase (Pck1), glucose-6-phosphatase (G6pc), collagen I (Col1a1), collagen III (Col3a1), fibronectin (Fn1), tissue inhibitor of metalloproteinase 1 (Timp1), and 2 (Timp2) were measured. T2D rats had higher serum glucose, cholesterol, LDL, and triglyceride levels compared to control. Liver Nr3c1, Col1a1, Col3a1, Fn1, Timp1, and Timp2 were increased in T2D rats. T2D liver showed significant fibrosis with the increases of α-smooth muscle actin and fibronectin. After gossypol treatment, serum glucose level was lowered by 64%. Liver fibrosis was significantly ameliorated. Nr3c1, Col1a1, Col3a1, Fn1, Timp1, Timp2, Pck1 as well as G6pc levels were significantly reduced. In conclusion, low dose gossypol is effective for the treatment of T2D and T2D-related fibrosis.
Collapse
Affiliation(s)
- Guorong Chen
- Institute of Cancer Research, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, PR China
| | - Rongrong Wang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Hanbin Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Liang Wu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Ren-Shan Ge
- Department of Anesthesiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Yili Wang
- Institute of Cancer Research, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, PR China.
| |
Collapse
|
22
|
You Y, Ren T, Zhang S, Shirima GG, Cheng Y, Liu X. Hypoglycemic effects of Zanthoxylum alkylamides by enhancing glucose metabolism and ameliorating pancreatic dysfunction in streptozotocin-induced diabetic rats. Food Funct 2015. [DOI: 10.1039/c5fo00432b] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alkylamides extracted from Zanthoxylum, a seasoning spice and folk medicine, exhibited hypoglycemic properties by enhancing glucose metabolism and ameliorating pancreatic dysfunction.
Collapse
Affiliation(s)
- Yuming You
- College of Food Science
- Southwest University
- Chongqing
- China
- College of Forestry and Life Science
| | - Ting Ren
- College of Food Science
- Southwest University
- Chongqing
- China
| | - Shiqi Zhang
- College of Food Science
- Southwest University
- Chongqing
- China
| | | | - YaJiao Cheng
- College of Food Science
- Southwest University
- Chongqing
- China
| | - Xiong Liu
- College of Food Science
- Southwest University
- Chongqing
- China
| |
Collapse
|
23
|
Karthi S, Manimaran P, Gandhimathi K, Ganesh R, Varalakshmi P, Ashokkumar B. Glucose-6-phosphatase (G6PC1) promoter polymorphism associated with glycogen storage disease type 1a among the Indian population. RSC Adv 2015. [DOI: 10.1039/c5ra10452a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Promoter polymorphism rs559748047 inG6PC1from GSD-1a among Indian population.
Collapse
Affiliation(s)
- Sellamuthu Karthi
- Department of Genetic Engineering
- School of Biotechnology
- Madurai Kamaraj University
- Madurai
- India
| | - Paramasivam Manimaran
- Department of Genetic Engineering
- School of Biotechnology
- Madurai Kamaraj University
- Madurai
- India
| | - Krishnan Gandhimathi
- Department of Genetic Engineering
- School of Biotechnology
- Madurai Kamaraj University
- Madurai
- India
| | - Ramasamy Ganesh
- Kanchi Kamakoti CHILDS Trust Hospital & The CHILDS Trust Medical Research Foundation
- Chennai
- India
| | - Perumal Varalakshmi
- Department of Molecular Microbiology
- School of Biotechnology
- Madurai Kamaraj University
- Madurai
- India
| | | |
Collapse
|
24
|
Insights into Transcriptional Regulation of Hepatic Glucose Production. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 318:203-53. [DOI: 10.1016/bs.ircmb.2015.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
25
|
Connor T, Martin SD, Howlett KF, McGee SL. Metabolic remodelling in obesity and type 2 diabetes: pathological or protective mechanisms in response to nutrient excess? Clin Exp Pharmacol Physiol 2014; 42:109-15. [DOI: 10.1111/1440-1681.12315] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 09/17/2014] [Accepted: 09/19/2014] [Indexed: 12/31/2022]
Affiliation(s)
- Timothy Connor
- Metabolic Remodelling Laboratory; Metabolic Research Unit; School of Medicine; Deakin University; Geelong Vic. Australia
| | - Sheree D Martin
- Metabolic Remodelling Laboratory; Metabolic Research Unit; School of Medicine; Deakin University; Geelong Vic. Australia
| | - Kirsten F Howlett
- Centre for Physical Activity and Nutrition; School of Exercise and Nutrition Sciences; Deakin University; Geelong Vic. Australia
| | - Sean L McGee
- Metabolic Remodelling Laboratory; Metabolic Research Unit; School of Medicine; Deakin University; Geelong Vic. Australia
- Division of Cell Signalling and Metabolism; Baker IDI Heart and Diabetes Institute; Melbourne Vic. Australia
| |
Collapse
|
26
|
Cho SJ, Park HJ, Jung UJ, Kim HJ, Moon BS, Choi MS. The beneficial effects of combined grape pomace and omija fruit extracts on hyperglycemia, adiposity and hepatic steatosis in db/db mice: a comparison with major index compounds. Int J Mol Sci 2014; 15:17778-89. [PMID: 25272231 PMCID: PMC4227189 DOI: 10.3390/ijms151017778] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/17/2014] [Accepted: 09/18/2014] [Indexed: 01/13/2023] Open
Abstract
This study investigated the effects of combined grape pomace and omija fruit extracts (GO) on diabetes-related metabolic changes in type 2 diabetic db/db mice. The effects of GO were compared with those of a resveratrol and schizandrin mixture (RS), which is a mixture of major components of GO. Mice were fed a normal diet with RS (0.005% resveratrol and 0.02% schizandrin in diet, w/w) or GO (0.3% grape pomace ethanol extract and 0.05% omija fruit ethanol extract in diet, w/w) for seven weeks. RS and GO not only lowered the levels of blood and plasma glucose, HbA1c, insulin and homeostasis model assessment of insulin resistance (HOMA-IR) with a simultaneous decrease in hepatic gluconeogenic enzymes activities and adiposity, but also improved preservation of the pancreatic β-cells. Plasma leptin and resistin levels were lower while the plasma adiponectin level was higher in the RS and GO groups than in the control group. Especially, GO increased hepatic glucokinase activity and gene expression and improved hepatic steatosis by elevating fatty acid oxidation compared to RS. These findings suggest that GO ameliorates hyperglycemia, adiposity and hepatic steatosis in type 2 diabetic mice.
Collapse
Affiliation(s)
- Su-Jung Cho
- Department of Food Science and Nutrition, Kyungpook National University, 1370 Sankyuk Dong Puk-ku, Daegu 702-701, Korea.
| | - Hae-Jin Park
- Department of Food Science and Nutrition, Kyungpook National University, 1370 Sankyuk Dong Puk-ku, Daegu 702-701, Korea.
| | - Un Ju Jung
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 1370 Sankyuk Dong Puk-ku, Daegu 702-701, Korea.
| | - Hye-Jin Kim
- Food Research & Development, CJ Cheiljedang Corporation, Seoul 152-051, Korea.
| | - Byoung Seok Moon
- Food Research & Development, CJ Cheiljedang Corporation, Seoul 152-051, Korea.
| | - Myung-Sook Choi
- Department of Food Science and Nutrition, Kyungpook National University, 1370 Sankyuk Dong Puk-ku, Daegu 702-701, Korea.
| |
Collapse
|
27
|
Keramati AR, Fathzadeh M, Go GW, Singh R, Choi M, Faramarzi S, Mane S, Kasaei M, Sarajzadeh-Fard K, Hwa J, Kidd KK, Babaee Bigi MA, Malekzadeh R, Hosseinian A, Babaei M, Lifton RP, Mani A. A form of the metabolic syndrome associated with mutations in DYRK1B. N Engl J Med 2014; 370:1909-1919. [PMID: 24827035 PMCID: PMC4069260 DOI: 10.1056/nejmoa1301824] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Genetic analysis has been successful in identifying causative mutations for individual cardiovascular risk factors. Success has been more limited in mapping susceptibility genes for clusters of cardiovascular risk traits, such as those in the metabolic syndrome. METHODS We identified three large families with coinheritance of early-onset coronary artery disease, central obesity, hypertension, and diabetes. We used linkage analysis and whole-exome sequencing to identify the disease-causing gene. RESULTS A founder mutation was identified in DYRK1B, substituting cysteine for arginine at position 102 in the highly conserved kinase-like domain. The mutation precisely cosegregated with the clinical syndrome in all the affected family members and was absent in unaffected family members and unrelated controls. Functional characterization of the disease gene revealed that nonmutant protein encoded by DYRK1B inhibits the SHH (sonic hedgehog) and Wnt signaling pathways and consequently enhances adipogenesis. Furthermore, DYRK1B promoted the expression of the key gluconeogenic enzyme glucose-6-phosphatase. The R102C allele showed gain-of-function activities by potentiating these effects. A second mutation, substituting proline for histidine 90, was found to cosegregate with a similar clinical syndrome in an ethnically distinct family. CONCLUSIONS These findings indicate a role for DYRK1B in adipogenesis and glucose homeostasis and associate its altered function with an inherited form of the metabolic syndrome. (Funded by the National Institutes of Health.).
Collapse
Affiliation(s)
- Ali R Keramati
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Mohsen Fathzadeh
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Gwang-Woong Go
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Rajvir Singh
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Murim Choi
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Saeed Faramarzi
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Shrikant Mane
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Mohammad Kasaei
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Kazem Sarajzadeh-Fard
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - John Hwa
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Kenneth K Kidd
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Mohammad A Babaee Bigi
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Reza Malekzadeh
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Adallat Hosseinian
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Masoud Babaei
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Richard P Lifton
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| | - Arya Mani
- Department of Internal Medicine, Yale Cardiovascular Research Center (A.R.K., M.F., G.-W.G., R.S., S.F., J.H., A.M.), Yale Center for Mendelian Genomics (M.C., S.M., R.P.L., A.M.), Department of Genetics (K.K.K., R.P.L., A.M.), and Howard Hughes Medical Institute (R.P.L.), Yale University School of Medicine, New Haven, CT; the Digestive Disease Research Institute, Shariati Hospital (M.F., K.S.-F., R.M.), and Department of Medical Genetics (M.F.), Tehran University of Medical Sciences, Tehran, the Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz (M.K., K.S.-F., M.A.B.B.), and Ardabil University of Medical Sciences, Ardabil (A.H., M.B.) - all in Iran
| |
Collapse
|
28
|
Abstract
Metabolic syndrome is defined by a constellation of interconnected physiological, biochemical, clinical, and metabolic factors that directly increases the risk of cardiovascular disease, type 2 diabetes mellitus, and all cause mortality. Insulin resistance, visceral adiposity, atherogenic dyslipidemia, endothelial dysfunction, genetic susceptibility, elevated blood pressure, hypercoagulable state, and chronic stress are the several factors which constitute the syndrome. Chronic inflammation is known to be associated with visceral obesity and insulin resistance which is characterized by production of abnormal adipocytokines such as tumor necrosis factor α , interleukin-1 (IL-1), IL-6, leptin, and adiponectin. The interaction between components of the clinical phenotype of the syndrome with its biological phenotype (insulin resistance, dyslipidemia, etc.) contributes to the development of a proinflammatory state and further a chronic, subclinical vascular inflammation which modulates and results in atherosclerotic processes. Lifestyle modification remains the initial intervention of choice for such population. Modern lifestyle modification therapy combines specific recommendations on diet and exercise with behavioural strategies. Pharmacological treatment should be considered for those whose risk factors are not adequately reduced with lifestyle changes. This review provides summary of literature related to the syndrome's definition, epidemiology, underlying pathogenesis, and treatment approaches of each of the risk factors comprising metabolic syndrome.
Collapse
Affiliation(s)
- Jaspinder Kaur
- Ex-Servicemen Contributory Health Scheme (ECHS) Polyclinic, Sultanpur Lodhi, Kapurthala District 144626, India
| |
Collapse
|
29
|
Das M, Banerjee B, Choudhury MG, Saha N. Environmental hypertonicity causes induction of gluconeogenesis in the air-breathing singhi catfish, Heteropneustes fossilis. PLoS One 2013; 8:e85535. [PMID: 24376888 PMCID: PMC3869940 DOI: 10.1371/journal.pone.0085535] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 11/27/2013] [Indexed: 02/01/2023] Open
Abstract
The air-breathing singhi catfish (Heteropneustes fossilis) is frequently being challenged by different environmental insults such as hyper-ammonia, dehydration and osmotic stresses in their natural habitats throughout the year. The present study investigated the effect of hyperosmotic stress, due to exposure to hypertonic environment (300 mM mannitol) for 14 days, on gluconeogenesis in this catfish. In situ exposure to hypertonic environment led to significant stimulation of gluconeogenic fluxes from the perfused liver after 7 days of exposure, followed by further increase after 14 days in presence of three different potential gluconeogenic substrates (lactate, pyruvate and glutamate). Environmental hypertonicity also caused a significant increase of activities of key gluconeogenic enzymes, namely phosphoenolpyruvate carboxykinase, fructose 1, 6-bisphosphatase and glucose 6-phosphatase by about 2-6 fold in liver, and 3-6 fold in kidney tissues. This was accompanied by more abundance of enzyme proteins by about 1.8–3.7 fold and mRNAs by about 2.2–5.2 fold in both the tissues with a maximum increase after 14 days of exposure. Hence, the increase in activities of key gluconeogenic enzymes under hypertonic stress appeared to be as a result of transcriptional regulation of genes. Immunocytochemical analysis further confirmed the tissue specific localized expression of these enzymes in both the tissues with the possibility of expressing more in the same localized places. The induction of gluconeogenesis during exposure to environmental hypertonicity possibly occurs as a consequence of changes in hydration status/cell volume of different cell types. Thus, these adaptational strategies related to gluconeogenesis that are observed in this catfish under hypertonic stress probably help in maintaining glucose homeostasis and also for a proper energy supply to support metabolic demands mainly for ion transport and other altered metabolic processes under various environmental hypertonic stress-related insults.
Collapse
Affiliation(s)
- Manas Das
- Biochemical Adaptation Laboratory, Department of Zoology, North-Eastern Hill University, Shillong, India
| | - Bodhisattwa Banerjee
- Biochemical Adaptation Laboratory, Department of Zoology, North-Eastern Hill University, Shillong, India
| | - Mahua G. Choudhury
- Biochemical Adaptation Laboratory, Department of Zoology, North-Eastern Hill University, Shillong, India
| | - Nirmalendu Saha
- Biochemical Adaptation Laboratory, Department of Zoology, North-Eastern Hill University, Shillong, India
- * E-mail:
| |
Collapse
|
30
|
Antidiabetic Activity of Polysaccharides from Tuberous Root of Liriope spicata var. prolifera in KKAy Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:349790. [PMID: 23762123 PMCID: PMC3677662 DOI: 10.1155/2013/349790] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 03/13/2013] [Accepted: 04/18/2013] [Indexed: 11/18/2022]
Abstract
Tuberous root of Liriope spicata var. prolifera has been widely used as a traditional Chinese medicine for the treatment of diabetes. The present study investigated the antidiabetic effect and the potential mechanisms of two new polysaccharides (LSP1, LSP2) and the total polysaccharides (TLSP), isolated from the tuberous roots. Upon the intragastric administration in obese insulin-resistant diabetic KKAy mice for 28 days, TLSP, LSP1, and LSP2 all caused a remarkable decrease of fasting blood glucose and significant improvement of insulin resistance and serum lipid metabolism in diabetic mice. In addition, liver histological analysis showed that TLSP, LSP1, and LSP2 significantly ameliorated the hepatocyte hypertrophy and decreased the lipid accumulation in the mice liver. Further experiments suggested that TLSP, LSP1, and LSP2 effectively inhibited hepatic gluconeogenesis and increased hepatic glycolysis and hepatic glycogen content. Furthermore, the mechanistic analysis showed the increased expression of insulin-receptor α subunit, insulin-receptor substrate-1, phosphatidylinositol 3-kinase, and peroxisome proliferators-activated receptors γ. These results suggested that TLSP, LSP1, and LSP2 manifest strong antidiabetic activity, therefore hold a great promise for therapeutic application in diabetic therapy and other related metabolic disorders.
Collapse
|
31
|
Huang PL, Chi CW, Liu TY. Areca nut procyanidins ameliorate streptozocin-induced hyperglycemia by regulating gluconeogenesis. Food Chem Toxicol 2013; 55:137-43. [PMID: 23333576 DOI: 10.1016/j.fct.2012.12.057] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 09/26/2012] [Accepted: 12/19/2012] [Indexed: 01/29/2023]
Abstract
Hepatic gluconeogenesis is a major contributor to blood glucose in diabetes mellitus. Our previous study indicated that areca nut extract enriched with catechin-based procyanidins from oligomers to polymers gave rise to anti-inflammatory effects in vitro and in vivo. Here we have surveyed the molecular features of areca nut procyanidins (ANPs) using quadrupole time-of-flight liquid chromatography/mass spectrometry (Q-TOF LC/MS) and the resulting mass spectrum accurately described ANP from monomer to hexadecamer. Furthermore, the potential of ANP in terms of blood glucose homeostasis was explored using cyclic adenosine monophosphate (cAMP)/dexamethasone stimulated primary mouse hepatocytes and multiple low dose streptozocin (MLD-STZ) treated mice. With the primary hepatocytes, ANP dose-dependently inhibited gluconeogenesis and reduced the mRNA expression of two gluconeogenic key enzymes, phosphoenol-pyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase). Intragastrically feeding of 10mg/kg ANP for 4weeks reduced the levels of fasting blood glucose, PEPCK and G6Pase in MLD-STZ mice. In additional, the level of 5'-AMP-activated protein kinase (AMPK) expression showed a trend towards being restored in the ANP treated MLD-STZ-mice. This study indicated that ANP has the potential to improve hyperglycemia by regulating gluconeogenic related kinases in MLD-STZ-mice.
Collapse
Affiliation(s)
- Pei-Ling Huang
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | |
Collapse
|
32
|
Kong LJ, Feng W, Wright M, Chen Y, Dallas-yang Q, Zhou YP, Berger JP. FGF21 suppresses hepatic glucose production through the activation of atypical protein kinase Cι/λ. Eur J Pharmacol 2013; 702:302-8. [PMID: 23305840 DOI: 10.1016/j.ejphar.2012.11.065] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 11/19/2012] [Accepted: 11/23/2012] [Indexed: 12/15/2022]
Abstract
Fibroblast growth factor 21 (FGF21) has been identified as a potent and robust metabolic regulator. Administration of recombinant FGF21 protein to rodents and rhesus monkeys exerts strong anti-diabetic effects. Previous studies have demonstrated that FGF21 inhibits glucose output in the rat H4IIE hepatoma cell line. We performed pharmacological studies to investigate the mechanisms by which FGF21 regulates glucose production in these cells. We found that both insulin and FGF21 suppressed gene expression of G6Pase and PEPCK. Accordingly, glucose production was inhibited. The FGF21 effects were phosphoinositide 3-kinase (PI3K)-dependent, and, unlike insulin, Akt-independent. Additionally, we found that FGF21 induced PKCι/λ phosphorylation in a PI3K-dependent manner; and that a non-isoform selective PKC inhibitor blocked FGF21 inhibition of glucose production, while an inhibitor of classical and novel PKC isoforms had no effect on FGF21 inhibitory activity. Furthermore, hepatic PKCι/λ phosphorylation was upregulated in FGF21-treated diabetic db/db mice.These data support the proposition that FGF21 inhibits hepatic glucose production by the PI3K-dependent activation of PKCι/λ.
Collapse
Affiliation(s)
- Ling-Jie Kong
- Merck Research Laboratories, RY80T-B119, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Despite the fact that insulin injection can protect diabetic patients from developing diabetes-related complications, recent meta-analyses indicate that rapid and long-acting insulin analogues only provide a limited benefit compared with conventional insulin regarding glycemic control. As insulin deficiency is the main sequel of type-1 diabetes (T1D), transfer of the insulin gene-by-gene therapy is becoming an attractive treatment modality even though T1D is not caused by a single genetic defect. In contrast to human insulin and insulin analogues, insulin gene therapy targets to supplement patients not only with insulin but also with C-peptide. So far, insulin gene therapy has had limited success because of delayed and/or transient gene expression. Sustained insulin gene expression is now feasible using current gene-therapy vectors providing patients with basal insulin coverage, but management of postprandial hyperglycaemia is still difficult to accomplish because of the inability to properly control insulin secretion. Enteroendocrine cells of the gastrointestinal track (K cells and L cells) may be ideal targets for insulin gene therapy, but cell-targeting difficulties have limited practical implementation of insulin gene therapy for diabetes treatment. Therefore, recent gene transfer technologies developed to generate authentic beta cells through transdifferentiation are also highlighted in this review.
Collapse
|
34
|
Esposito D, Kizelsztein P, Komarnytsky S, Raskin I. Hypoglycemic effects of brassinosteroid in diet-induced obese mice. Am J Physiol Endocrinol Metab 2012; 303:E652-8. [PMID: 22785239 PMCID: PMC3774328 DOI: 10.1152/ajpendo.00024.2012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The prevalence of obesity is increasing globally, and obesity is a major risk factor for metabolic diseases such as type 2 diabetes. Previously, we reported that oral administration of homobrassinolide (HB) to healthy rats triggered a selective anabolic response that was associated with lower blood glucose. Therefore, the aim of this study was to evaluate the effects of HB administration on glucose metabolism, insulin sensitivity, body composition, and gluconeogenic gene expression profiles in liver of C57BL/6J high-fat diet-induced obese mice. Acute oral administration of 50-300 mg/kg HB to obese mice resulted in a dose-dependent decrease in fasting blood glucose within 3 h of treatment. Daily chronic administration of HB (50 mg/kg for 8 wk) ameliorated hyperglycemia and improved oral glucose tolerance associated with obesity without significantly affecting body weight or body composition. These changes were accompanied by lower expression of two key gluconeogenic enzymes, phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G-6-Pase), and increased phosphorylation of AMP-activated protein kinase in the liver and muscle tissue. In vitro, HB treatment (1-15 μM) inhibited cyclic AMP-stimulated but not dexamethasone-stimulated upregulation of PEPCK and G-6-Pase mRNA levels in H4IIE rat hepatoma cells. Among a series of brassinosteroid analogs related to HB, only homocastasterone decreased glucose production in cell culture significantly. These results indicate the antidiabetic effects of brassinosteroids and begin to elucidate their putative cellular targets both in vitro and in vivo.
Collapse
Affiliation(s)
- Debora Esposito
- Biotech Center, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | | | | | | |
Collapse
|
35
|
Jia Y, Cong R, Li R, Yang X, Sun Q, Parvizi N, Zhao R. Maternal low-protein diet induces gender-dependent changes in epigenetic regulation of the glucose-6-phosphatase gene in newborn piglet liver. J Nutr 2012; 142:1659-65. [PMID: 22833655 DOI: 10.3945/jn.112.160341] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Glucose-6-phosphatase (G6PC) plays an important role in glucose homeostasis because it catalyzes the final steps of gluconeogenesis and glycogenolysis. Maternal malnutrition during pregnancy affects G6PC activity, yet it is unknown whether epigenetic regulations of the G6PC gene are also affected. In this study, we fed primiparous, purebred Meishan sows either standard-protein (SP; 12% crude protein) or low-protein (LP; 6% crude protein) diets throughout gestation and analyzed hepatic G6PC expression in both male and female newborn piglets. The epigenetic regulation of G6PC, including DNA methylation, histone modifications, and micro RNA (miRNA), was determined to reveal potential mechanisms. Male, but not female, LP piglets had a significantly lower serum glucose concentration and greater hepatic G6PC mRNA expression and enzyme activity. Also, in LP males, glucocorticoid receptor binding to the G6PC promoter was lower compared with SP males, which was accompanied by hypomethylation of the G6PC promoter. Modifications in histones also were gender dependent; LP males had less histone H3 and histone H3 lysine 9 trimethylation and more histone H3 acetylation and histone H3 lysine 4 trimethylation on the G6PC promoter compared with the SP males, whereas LP females had more H3 and greater H3 methylation compared with their SP counterparts. Moreover, two miRNA, ssc-miR-339-5p and ssc-miR-532-3p, targeting the G6PC 3' untranslated region were significantly upregulated by the LP diet only in females. These results suggest that a maternal LP diet during pregnancy causes hepatic activation of G6PC gene expression in male piglets, which possibly contributes to adult-onset hyperglycemia.
Collapse
Affiliation(s)
- Yimin Jia
- Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, PR China
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Insulin resistance with its associated hyperglycemias represents one significant contributor to mortality in burned patients. A variety of cellular stress-signaling pathways are activated as a consequence of burn. A key player in the cellular stress response is the endoplasmic reticulum (ER). Here, we investigated a possible role for ER-stress pathways in the progression of insulin function dysregulation postburn. Rats received a 60% total body surface area thermal injury, and a laparotomy was performed at 24, 72, and 192 h postburn. Liver was harvested before and 1 min after insulin injection (1 IU/kg) into the portal vein, and expression patterns of various proteins known to be involved in insulin and ER-stress signaling were determined by Western blotting. mRNA expression of glucose-6-phosphatase and glucokinase were determined by reverse-transcriptase-polymerase chain reaction and fasting serum glucose and insulin levels by standard enzymatic and enzyme-linked immunosorbent assay techniques, respectively. Insulin resistance indicated by increased glucose and insulin levels occurred starting 24 h postburn. Burn injury resulted in activation of ER stress pathways, reflected by significantly increased accumulation of phospho-PKR-like ER-kinase and phosphorylated inositol requiring enzyme 1, leading to an elevation of phospho-c-Jun N-terminal kinase and serine phosphorylation of insulin receptor substrate (IRS) 1 postburn. Insulin administration caused a significant increase in tyrosine phosphorylation of IRS-1, leading to activation of the phosphatidylinositol 3 kinase/Akt pathway in normal liver. Postburn tyrosine phosphorylation of IRS-1 was significantly impaired, associated with an inactivation of signaling molecules acting downstream of IRS-1, leading to significantly elevated transcription of glucose-6-phosphatase and significantly decreased mRNA expression of glucokinase. Activation of ER-stress signaling cascades may explain metabolic abnormalities involving insulin action after burn.
Collapse
|
37
|
Konopelska S, Kienitz T, Quinkler M. Downregulation of hepatic glucose-6-phosphatase-α in patients with hepatic steatosis. Obesity (Silver Spring) 2011; 19:2322-6. [PMID: 21593806 DOI: 10.1038/oby.2011.118] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Glucose-6-phosphate transporter (G6PT) and microsomal glucose-6-phosphatase-α (G6Pase-α) perform the terminal step in glycogenolysis and gluconeogenesis. Deficiency of these proteins leads to glycogen storage diseases. Partial inhibition of G6Pase in rats results in increased hepatic triglyceride content and de novo lipogenesis leading to hepatic steatosis. Hepatic steatosis represents hepatic manifestation of the metabolic syndrome. We investigated molecular mechanisms that may explain the relationship between fatty liver and G6Pase-α in humans in detail. A total of 27 patients (11 men, 16 women) underwent liver biopsy. Histological diagnosis identified nonfatty liver in seven patients and nonalcoholic fatty liver in 20 patients. We quantified G6Pase-α and G6PT mRNA expression by real-time PCR. Anthropometric measurements and analysis of plasma lipids and liver enzymes were performed. Patients with fatty liver showed no significant differences in age, HOMA(IR) (homeostasis model assessment of insulin resistance), BMI, liver enzymes or waist-to-hip ratio compared to those with nonfatty liver, but total plasma cholesterol levels and liver fat content were higher in patients with fatty liver (P < 0.05). G6Pase-α and G6PT mRNA expressions were significantly downregulated in fatty compared to histologically normal liver (P < 0.05). G6Pase-α and G6PT mRNA expressions correlated positively (R(2) = 0.406 P < 0.05). Both expressions did not correlate with age, BMI, aspartate transaminase, alanine transaminase, alkaline phosphatase, γ-glutamyl transferase, triglycerides or glucose levels. Our data suggest that expression of hepatic G6Pase-α and G6PT are closely interlinked. Downregulation of G6Pase-α in fatty liver might be associated with hepatic fat accumulation and pathogenesis of hepatic steatosis.
Collapse
Affiliation(s)
- Sarah Konopelska
- Department of Clinical Endocrinology, Charité Campus Mitte, Charité University Medicine Berlin, Berlin, Germany.
| | | | | |
Collapse
|
38
|
Shabrova EV, Tarnopolsky O, Singh AP, Plutzky J, Vorsa N, Quadro L. Insights into the molecular mechanisms of the anti-atherogenic actions of flavonoids in normal and obese mice. PLoS One 2011; 6:e24634. [PMID: 22016761 PMCID: PMC3189911 DOI: 10.1371/journal.pone.0024634] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 08/17/2011] [Indexed: 01/02/2023] Open
Abstract
Obesity is a major and independent risk factor for cardiovascular disease and it is strongly associated with the development of dyslipidemia, insulin resistance and type 2 diabetes. Flavonoids, a diverse group of polyphenol compounds of plant origin widely distributed in human diet, have been reported to have numerous health benefits, although the mechanisms underlying these effects have remained obscure. We analyzed the effects of chronic dietary supplementation with flavonoids extracted from cranberry (FLS) in normal and obese C57/BL6 mice compared to mice maintained on the same diets lacking FLS. Obese mice supplemented with flavonoids showed an amelioration of insulin resistance and plasma lipid profile, and a reduction of visceral fat mass. We provide evidence that the adiponectin-AMPK pathway is the main mediator of the improvement of these metabolic disorders. In contrast, the reduced plasma atherogenic cholesterol observed in normal mice under FLS seems to be due to a downregulation of the hepatic cholesterol synthesis pathway. Overall, we demonstrate for the first time that the molecular mechanisms underlying the beneficial effects of flavonoids are determined by the metabolic state.
Collapse
Affiliation(s)
- Elena V. Shabrova
- Department of Plant Biology and Pathology, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
- Food Science Department and Rutgers Center for Lipid Research, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Olga Tarnopolsky
- Department of Plant Biology and Pathology, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Ajay P. Singh
- Department of Plant Biology and Pathology, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Jorge Plutzky
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts, United States of America
| | - Nicholi Vorsa
- Department of Plant Biology and Pathology, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
- PE Marucci Center, Rutgers, The State University of New Jersey, Chatsworth, New Jersey, United States of America
- * E-mail: (LQ); (NV)
| | - Loredana Quadro
- Food Science Department and Rutgers Center for Lipid Research, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
- * E-mail: (LQ); (NV)
| |
Collapse
|
39
|
Wan M, Leavens KF, Saleh D, Easton RM, Guertin DA, Peterson TR, Kaestner KH, Sabatini DM, Birnbaum MJ. Postprandial hepatic lipid metabolism requires signaling through Akt2 independent of the transcription factors FoxA2, FoxO1, and SREBP1c. Cell Metab 2011; 14:516-27. [PMID: 21982711 PMCID: PMC3190164 DOI: 10.1016/j.cmet.2011.09.001] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 06/13/2011] [Accepted: 09/01/2011] [Indexed: 12/17/2022]
Abstract
Under conditions of obesity and insulin resistance, the serine/threonine protein kinase Akt/PKB is required for lipid accumulation in liver. Two forkhead transcription factors, FoxA2 and FoxO1, have been suggested to function downstream of and to be negatively regulated by Akt and are proposed as key determinants of hepatic triglyceride content. In this study, we utilize genetic loss of function experiments to show that constitutive activation of neither FoxA2 nor FoxO1 can account for the protection from steatosis afforded by deletion of Akt2 in liver. Rather, another downstream target positively regulated by Akt, the mTORC1 complex, is required in vivo for de novo lipogenesis and Srebp1c expression. Nonetheless, activation of mTORC1 and SREBP1c is not sufficient to drive postprandial lipogenesis in the absence of Akt2. These data show that insulin signaling through Akt2 promotes anabolic lipid metabolism independent of Foxa2 or FoxO1 and through pathways additional to the mTORC1-dependent activation of SREBP1c.
Collapse
Affiliation(s)
- Min Wan
- The Institute of Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Im SS, Kim MY, Kwon SK, Kim TH, Bae JS, Kim H, Kim KS, Oh GT, Ahn YH. Peroxisome proliferator-activated receptor {alpha} is responsible for the up-regulation of hepatic glucose-6-phosphatase gene expression in fasting and db/db Mice. J Biol Chem 2011; 286:1157-1164. [PMID: 21081500 PMCID: PMC3020722 DOI: 10.1074/jbc.m110.157875] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 10/25/2010] [Indexed: 12/13/2022] Open
Abstract
Glucose-6-phosphatase (G6Pase) is a key enzyme that is responsible for the production of glucose in the liver during fasting or in type 2 diabetes mellitus (T2DM). During fasting or in T2DM, peroxisome proliferator-activated receptor α (PPARα) is activated, which may contribute to increased hepatic glucose output. However, the mechanism by which PPARα up-regulates hepatic G6Pase gene expression in these states is not well understood. We evaluated the mechanism by which PPARα up-regulates hepatic G6Pase gene expression in fasting and T2DM states. In PPARα-null mice, both hepatic G6Pase and phosphoenolpyruvate carboxykinase levels were not increased in the fasting state. Moreover, treatment of primary cultured hepatocytes with Wy14,643 or fenofibrate increased the G6Pase mRNA level. In addition, we have localized and characterized a PPAR-responsive element in the promoter region of the G6Pase gene. Chromatin immunoprecipitation (ChIP) assay revealed that PPARα binding to the putative PPAR-responsive element of the G6Pase promoter was increased in fasted wild-type mice and db/db mice. These results indicate that PPARα is responsible for glucose production through the up-regulation of hepatic G6Pase gene expression during fasting or T2DM animal models.
Collapse
Affiliation(s)
- Seung-Soon Im
- From the Department of Biochemistry and Molecular Biology
- the Center for Chronic Metabolic Disease Research, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
- the Metabolic Signaling and Disease Program, Sanford-Burnham Medical Research Institute, Orlando, Florida 32827, and
| | - Mi-Young Kim
- From the Department of Biochemistry and Molecular Biology
- the Center for Chronic Metabolic Disease Research, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Sool-Ki Kwon
- From the Department of Biochemistry and Molecular Biology
- the Center for Chronic Metabolic Disease Research, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Tae-Hyun Kim
- From the Department of Biochemistry and Molecular Biology
- the Brain Korea 21 Project for Medical Sciences, and
- the Center for Chronic Metabolic Disease Research, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Jin-Sik Bae
- From the Department of Biochemistry and Molecular Biology
- the Center for Chronic Metabolic Disease Research, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Hail Kim
- From the Department of Biochemistry and Molecular Biology
- the Center for Chronic Metabolic Disease Research, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Kyung-Sup Kim
- From the Department of Biochemistry and Molecular Biology
- the Brain Korea 21 Project for Medical Sciences, and
- the Center for Chronic Metabolic Disease Research, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Goo-Taeg Oh
- the Laboratory of Cardiovascular Genomics, Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Yong-Ho Ahn
- From the Department of Biochemistry and Molecular Biology
- the Brain Korea 21 Project for Medical Sciences, and
- the Center for Chronic Metabolic Disease Research, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| |
Collapse
|
41
|
Gregory JM, Moore DJ. Can technological solutions for diabetes replace islet cell function? Organogenesis 2011; 7:32-41. [PMID: 21289480 DOI: 10.4161/org.7.1.14028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The central objective of diabetes research and management is to restore the deficient secretion of insulin, thereby restoring a state of euglycemia and minimizing short- and long-term risks associated with poor glucose control. The development of the artificial pancreas seeks to imitate the action of the pancreatic beta cell by employing closed-loop control to respond to glycemic excursions by appropriately infusing appropriate amounts of insulin. This article examines progress towards implementing an artificial pancreas in the context of the pancreatic islet as the ideal model for controlling blood glucose. Physiologic insulin secretion will form our foundation for considering the technical design elements relevant to electromechanically imitating the beta cell. The most recent clinical trials using closed-loop control are reviewed and this modality is compared to other curative approaches including islet cell transplantation and preservation. Finally, the potential of the artificial pancreas as a method to adequately reestablish euglycemia is considered.
Collapse
Affiliation(s)
- Justin M Gregory
- Department of Pediatrics, University of Tennessee School of Medicine, Memphis, TN, USA
| | | |
Collapse
|
42
|
Semecarpus anacardium (Bhallataka) Alters the Glucose Metabolism and Energy Production in Diabetic Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2010; 2011. [PMID: 20924498 PMCID: PMC2949585 DOI: 10.1155/2011/142978] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Revised: 05/11/2010] [Accepted: 06/27/2010] [Indexed: 01/19/2023]
Abstract
Glucose produced by gluconeogenesis and glycogenolysis plays an important role in aggravating hyperglycemia in diabetes, and altered mitochondrial function is associated with impaired energy production. The present study focuses on the effect of Semecarpus anacardium on carbohydrate metabolism and energy production in diabetic rats. Diabetes was induced by the administration of Streptozotocin at a dose of 50 mg/kg.b.wt. Three days after the induction, Semecarpus anacardium at a dose of 300 mg/kg.b.wt was administered for 21 days. After the experimental duration, the activities of the enzymes involved in Glycolysis, TCA cycle, gluconeogenesis, and glycogen were assayed in the liver and kidney of the experimental animals. In addition, to the complexes the protein expression of AKT and PI3K were assayed. The levels of the enzymes involved in Glycolysis and TCA cycle increased, while that of gluconeogensis decreased. The activities of the mitochondrial complexes were also favorably modulated. The expressions of PI3K and AKT also increased in the skeletal muscle. These effects may be attributed to the hypoglycemic and the antioxidative activity of Semecarpus anacardium. The results of the study revealed that Semecarpus anacardium was able to restore the altered activities of the enzymes involved in carbohydrate metabolism and energy production.
Collapse
|
43
|
Anti-diabetic effects of lemon balm (Melissa officinalis) essential oil on glucose- and lipid-regulating enzymes in type 2 diabetic mice. Br J Nutr 2010; 104:180-8. [DOI: 10.1017/s0007114510001765] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The antioxidant activity of lemon balm (Melissa officinalis) essential oil (LBEO) on 2,2-diphenyl-1-picrylhydrazyl (DPPH) radicals and its hypoglycaemic effect in db/db mice were investigated. LBEO scavenged 97 % of DPPH radicals at a 270-fold dilution. Mice administered LBEO (0·015 mg/d) for 6 weeks showed significantly reduced blood glucose (65 %; P < 0·05) and TAG concentrations, improved glucose tolerance, as assessed by an oral glucose tolerance test, and significantly higher serum insulin levels, compared with the control group. The hypoglycaemic mechanism of LBEO was further explored via gene and protein expression analyses using RT-PCR and Western blotting, respectively. Among all glucose metabolism-related genes studied, hepatic glucokinase and GLUT4, as well as adipocyte GLUT4, PPAR-γ, PPAR-α and SREBP-1c expression, were significantly up-regulated, whereas glucose-6-phosphatase and phosphoenolpyruvate carboxykinase expression was down-regulated in the livers of the LBEO group. The results further suggest that LBEO administered at low concentrations is an efficient hypoglycaemic agent, probably due to enhanced glucose uptake and metabolism in the liver and adipose tissue and the inhibition of gluconeogenesis in the liver.
Collapse
|
44
|
Fasting hyperglycemia is not associated with increased expression of PEPCK or G6Pc in patients with Type 2 Diabetes. Proc Natl Acad Sci U S A 2009; 106:12121-6. [PMID: 19587243 DOI: 10.1073/pnas.0812547106] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fasting hyperglycemia in patients with type 2 diabetes mellitus (T2DM) is attributed to increased hepatic gluconeogenesis, which has been ascribed to increased transcriptional expression of phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase, catalytic (G6Pc). To test this hypothesis, we examined hepatic expression of these 2 key gluconeogenic enzymes in 2 rodent models of fasting hyperglycemia and in patients with T2DM. In rats, high-fat feeding (HFF) induces insulin resistance but a robust beta-cell response prevents hyperglycemia. Fasting hyperglycemia was induced in the first rat model by using nicotinamide and streptozotocin to prevent beta-cell compensation, in combination with HFF (STZ/HFF). In a second model, control and HFF rats were infused with somatostatin, followed by portal vein infusion of insulin and glucagon. Finally, the expression of these enzymes was measured in liver biopsy samples obtained from insulin sensitive, insulin resistant, and untreated T2DM patients undergoing bariatric surgery. Rats treated with STZ/HFF developed modest fasting hyperglycemia (119 +/- 4 vs. 153 +/- 6 mg/dL, P < 0.001) and increased rates of endogenous glucose production (EGP) (4.6 +/- 0.6 vs. 6.9 +/- 0.6 mg/kg/min, P = 0.02). Surprisingly, the expression of PEPCK or G6Pc was not increased. Matching plasma insulin and glucagon with portal infusions led to higher plasma glucoses in the HFF rats (147 +/- 4 vs. 161 +/- 4 mg/dL, P = 0.05) with higher rates of EGP and gluconeogenesis. However, PEPCK and G6Pc expression remained unchanged. Finally, in patients with T2DM, hepatic expression of PEPCK or G6Pc was not increased. Thus, in contrast to current dogma, these data demonstrate that increased transcriptional expression of PEPCK1 and G6Pc does not account for increased gluconeogenesis and fasting hyperglycemia in patients with T2DM.
Collapse
|
45
|
Kim HI, Koh YK, Kim TH, Kwon SK, Im SS, Choi HS, Kim KS, Ahn YH. Transcriptional activation of SHP by PPAR-gamma in liver. Biochem Biophys Res Commun 2007; 360:301-306. [PMID: 17601490 DOI: 10.1016/j.bbrc.2007.05.171] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Accepted: 05/18/2007] [Indexed: 10/23/2022]
Abstract
The mechanism of how PPARgamma decrease gluconeogenic gene expressions in liver is still unclear. Since PPARgamma is a transcriptional activator, it requires a mediator to decrease the transcription of gluconeogenic genes. Recently, SHP has been shown to mediate the bile acid-dependent down regulation of gluconeogenic gene expression in liver. This led us to explore the possibility that SHP may mediate the antigluconeogenic effect of PPARgamma. In the present study, we have identified and characterized the presence of functional PPRE in human SHP promoter. We show the binding of PPARgamma/RXRalpha heterodimer to the PPRE and increased SHP expression by rosiglitazone in primary rat hepatocytes. Taken together with the previous reports about the function of SHP on gluconeogenesis, our results indicate that SHP can mediate the acute antigluconeogenic effect of PPARgamma.
Collapse
Affiliation(s)
- Ha-il Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemoon-gu, Seoul 120-752, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Qi D, Rodrigues B. Glucocorticoids produce whole body insulin resistance with changes in cardiac metabolism. Am J Physiol Endocrinol Metab 2007; 292:E654-67. [PMID: 17077342 DOI: 10.1152/ajpendo.00453.2006] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Insulin resistance is viewed as an insufficiency in insulin action, with glucocorticoids being recognized to play a key role in its pathogenesis. With insulin resistance, metabolism in multiple organ systems such as skeletal muscle, liver, and adipose tissue is altered. These metabolic alterations are widely believed to be important factors in the morbidity and mortality of cardiovascular disease. More importantly, clinical and experimental studies have established that metabolic abnormalities in the heart per se also play a crucial role in the development of heart failure. Following glucocorticoids, glucose utilization is compromised in the heart. This attenuated glucose metabolism is associated with altered fatty acid supply, composition, and utilization. In the heart, elevated fatty acid use has been implicated in a number of metabolic, morphological, and mechanical changes and, more recently, in "lipotoxicity". In the present article, we review the action of glucocorticoids, their role in insulin resistance, and their influence in modulating peripheral and cardiac metabolism and heart disease.
Collapse
Affiliation(s)
- Dake Qi
- Division of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, The University of British Columbia, 2146 East Mall, Vancouver, BC, Canada V6T 1Z3
| | | |
Collapse
|
47
|
Xu C, Chakravarty K, Kong X, Tuy TT, Arinze IJ, Bone F, Massillon D. Several transcription factors are recruited to the glucose-6-phosphatase gene promoter in response to palmitate in rat hepatocytes and H4IIE cells. J Nutr 2007; 137:554-9. [PMID: 17311939 DOI: 10.1093/jn/137.3.554] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Fatty acids and glucose are strong modulators of the expression of glucose-6-phosphatase (Glc-6-Pase), an enzyme that plays a key role in glucose homeostasis. PUFA inhibit, whereas SFA and monounsaturated fatty acids induce the expression of the Glc-6-Pase gene. Palmitate and oleate are the most abundant fatty acid species in circulation during food deprivation in mammals. Although dietary fats have been shown to modulate the expression of genes involved in both lipid and carbohydrate metabolism in liver, little is known regarding the molecular mechanism of transcriptional response of the Glc-6-Pase gene to long-chain fatty acids. Using H4IIE hepatoma cells and hepatocytes from adult rats, we investigated the mechanism of the induction of this gene by palmitate and oleate. Both of these fatty acids stimulated Glc-6-Pase gene transcription but did not affect the stability of its mRNA. In transient transfection assays, transcription from the Glc-6-Pase gene promoter was markedly enhanced by both palmitate and oleate but not by arachidonate. Chromatin immunoprecipitation analysis was used to show that palmitate induced the recruitment of an array of transcription factors viz hepatic nuclear factor(NF)-4alpha, CAAT/enhancer binding proteinbeta, PPARalpha, chicken ovalbumin upstream promoter transcription factor (COUP-TF), cAMP regulatory element binding protein, and NF-kappaB to this gene promoter. Although it is presently unclear how these various transcription factors interact at this promoter, the data are consistent with the view that multiple regulatory elements in the Glc-6-Pase gene promoter are responsible for the modulation of gene transcription by fatty acids.
Collapse
Affiliation(s)
- Chuan Xu
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH, 44106-4935, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Pedersen KB, Zhang P, Doumen C, Charbonnet M, Lu D, Newgard CB, Haycock JW, Lange AJ, Scott DK. The promoter for the gene encoding the catalytic subunit of rat glucose-6-phosphatase contains two distinct glucose-responsive regions. Am J Physiol Endocrinol Metab 2007; 292:E788-801. [PMID: 17106062 DOI: 10.1152/ajpendo.00510.2006] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glucose homeostasis requires the proper expression and regulation of the catalytic subunit of glucose-6-phosphatase (G-6-Pase), which hydrolyzes glucose 6-phosphate to glucose in glucose-producing tissues. Glucose induces the expression of G-6-Pase at the transcriptional and posttranscriptional levels by unknown mechanisms. To better understand this metabolic regulation, we mapped the cis-regulatory elements conferring glucose responsiveness to the rat G-6-Pase gene promoter in glucose-responsive cell lines. The full-length (-4078/+64) promoter conferred a moderate glucose response to a reporter construct in HL1C rat hepatoma cells, which was dependent on coexpression of glucokinase. The same construct provided a robust glucose response in 832/13 INS-1 rat insulinoma cells, which are not glucogenic. Glucose also strongly increased endogenous G-6-Pase mRNA levels in 832/13 cells and in rat pancreatic islets, although the induced levels from islets were still markedly lower than in untreated primary hepatocytes. A distal promoter region was glucose responsive in 832/13 cells and contained a carbohydrate response element with two E-boxes separated by five base pairs. Carbohydrate response element-binding protein bound this region in a glucose-dependent manner in situ. A second, proximal promoter region was glucose responsive in both 832/13 and HL1C cells, with a hepatocyte nuclear factor 1 binding site and two cAMP response elements required for glucose responsiveness. Expression of dominant-negative versions of both cAMP response element-binding protein and CAAT/enhancer-binding protein blocked the glucose response of the proximal region in a dose-dependent manner. We conclude that multiple, distinct cis-regulatory promoter elements are involved in the glucose response of the rat G-6-Pase gene.
Collapse
Affiliation(s)
- Kim B Pedersen
- Department of Biochemistry and Molecular Biology, Lousiana State University Health Sciences Center, New Orleans, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lan MS, Wang HW, Chong J, Breslin MB. Coupling of glucose response element from L-type pyruvate kinase and G6Pase promoter enhances glucose responsive activity in hepatoma cells. Mol Cell Biochem 2006; 300:191-6. [PMID: 17160355 DOI: 10.1007/s11010-006-9383-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Accepted: 11/07/2006] [Indexed: 11/24/2022]
Abstract
Type 1 diabetes results from the autoimmune destruction of pancreatic beta-cells, which leads to severe insulin deficiency. Insulin gene therapy provides an attractive approach to cure diabetes. The critical factor for insulin gene therapy in surrogate cells is to select an appropriate site for insulin expression and a tissue-specific promoter that is responsive to both physiological glucose and insulin concentrations. A novel chimeric promoter, (GIRE)n-G6Pase, consisting of a 1.6 kb glucose 6-phosphatase (G6Pase) promoter and a segment of the regulatory element derived from the L-type pyruvate kinase (L-PK) promoter, was designed to provide strong and tight control of insulin expression in liver. One or three copies of GIRE were linked to the G6Pase promoter, which showed a stronger promoter activity than the G6Pase promoter alone. The chimeric promoter was inhibited by insulin in a dosage-dependent manner and activated by glucose, two features essential for glucose metabolism. The promoter activity is conserved between species and highly specific for liver cells. The construction of a chimeric promoter with stronger and more sensitive responsive activity to glucose and insulin in liver cells could further advance studies in insulin gene therapy.
Collapse
Affiliation(s)
- Michael S Lan
- The Research Institute for Children, Departments of Pediatrics and Genetics, Children's Hospital, Louisiana State University Health Sciences Center, 200 Henry Clay Avenue, New Orleans, LA 70118, USA.
| | | | | | | |
Collapse
|
50
|
Hall RK, Wang XL, George L, Koch SR, Granner DK. Insulin represses phosphoenolpyruvate carboxykinase gene transcription by causing the rapid disruption of an active transcription complex: a potential epigenetic effect. Mol Endocrinol 2006; 21:550-63. [PMID: 17095578 DOI: 10.1210/me.2006-0307] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Insulin represses gluconeogenesis, in part, by inhibiting the transcription of genes that encode rate-determining enzymes, such as phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G-6-Pase). Glucocorticoids stimulate expression of the PEPCK gene but the repressive action of insulin is dominant. Here, we show that treatment of H4IIE hepatoma cells with the synthetic glucocorticoid, dexamethasone (dex), induces the accumulation of glucocorticoid receptor, as well as many transcription factors, coregulators, and RNA polymerase II, on the PEPCK gene promoter. The addition of insulin to dex-treated cells causes the rapid dissociation of glucocorticoid receptor, polymerase II, and several key transcriptional regulators from the PEPCK gene promoter. These changes are temporally related to the reduced rate of PEPCK gene transcription. A similar disruption of the G-6-Pase gene transcription complex was observed. Additionally, insulin causes the rapid demethylation of arginine-17 on histone H3 of both genes. This rapid, insulin-induced, histone demethylation is temporally related to the disruption of the PEPCK and G-6-Pase gene transcription complex, and may be causally related to the mechanism by which insulin represses transcription of these genes.
Collapse
Affiliation(s)
- Robert K Hall
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 707 Light Hall, Nashville, Tennessee 37232-0615, USA
| | | | | | | | | |
Collapse
|