151
|
Ocłoń E, Latacz A, Zubel-Łojek J, Pierzchała-Koziec K. Hyperglycemia-induced changes in miRNA expression patterns in epicardial adipose tissue of piglets. J Endocrinol 2016; 229:259-66. [PMID: 27044779 DOI: 10.1530/joe-15-0495] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 03/30/2016] [Indexed: 12/28/2022]
Abstract
MicroRNAs (miRNAs) are a class of molecular posttranscriptional regulators found to participate in numerous biological mechanisms, such as adipogenesis, fat deposition, or glucose metabolism. Additionally, a detailed analysis on the molecular and cellular mechanisms of miRNA-related effects on metabolism leads to developing novel diagnostic markers and therapeutic approaches. To identify miRNA whose activity changed in epicardial adipose tissue in piglets during hyperglycemia, we analyzed the different miRNA expression patterns between control and hyperglycemia groups. The microarray analysis selected three differentially expressed microRNAs as potential biomarkers: hsa-miR-675-5p, ssc-miR-193a-3p, and hsa-miR-144-3p. The validation of miRNA expression with real-time PCR indicated an increased expression levels of ssc-miR-193a-3p and miR-675-5p, whereas the expression level of hsa-miR-144-3p was lower in epicardial adipose tissue in response to hyperglycemia (P<0.01). The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses suggested that these miRNAs differentially expressed between hyperglycemic and control piglets are involved in insulin, adipocytokine, and phosphatidylinositol 3-kinase-Akt signaling pathways, and development of type 2 diabetes as well. The results suggested that hyperglycemia can significantly affect the expression patterns of miRNA in porcine adipose tissue.
Collapse
Affiliation(s)
- Ewa Ocłoń
- Department of Animal Physiology and EndocrinologyUniversity of Agriculture in Krakow, Krakow, Poland
| | - Anna Latacz
- Department of Animal Physiology and EndocrinologyUniversity of Agriculture in Krakow, Krakow, Poland
| | - Joanna Zubel-Łojek
- Department of Animal Physiology and EndocrinologyUniversity of Agriculture in Krakow, Krakow, Poland
| | | |
Collapse
|
152
|
Hinshaw L, Schiavon M, Dadlani V, Mallad A, Dalla Man C, Bharucha A, Basu R, Geske JR, Carter RE, Cobelli C, Basu A, Kudva YC. Effect of Pramlintide on Postprandial Glucose Fluxes in Type 1 Diabetes. J Clin Endocrinol Metab 2016; 101:1954-62. [PMID: 26930181 PMCID: PMC4870844 DOI: 10.1210/jc.2015-3952] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CONTEXT Early postprandial hyperglycemia and delayed hypoglycemia remain major problems in current management of type 1 diabetes (T1D). OBJECTIVE Our objective was to investigate the effects of pramlintide, known to suppress glucagon and delay gastric emptying, on postprandial glucose fluxes in T1D. DESIGN This was a single-center, inpatient, randomized, crossover study. PATIENTS Twelve patients with T1D who completed the study were analyzed. INTERVENTIONS Subjects were studied on two occasions with or without pramlintide. Triple tracer mixed-meal method and oral minimal model were used to estimate postprandial glucose turnover and insulin sensitivity (SI). Integrated liver insulin sensitivity was calculated based on glucose turnover. Plasma glucagon and insulin were measured. MAIN OUTCOME MEASURE Glucose turnover and SI were the main outcome measures. RESULTS With pramlintide, 2-hour postprandial glucose, insulin, glucagon, glucose turnover, and SI indices showed: plasma glucose excursions were reduced (difference in incremental area under the curve [iAUC], 444.0 mMmin, P = .0003); plasma insulin concentrations were lower (difference in iAUC, 7642.0 pMmin; P = .0099); plasma glucagon excursions were lower (difference in iAUC, 1730.6 pg/mlmin; P = .0147); meal rate of glucose appearance was lower (difference in iAUC: 1196.2 μM/kg fat free mass [FFM]; P = .0316), endogenous glucose production was not different (difference in iAUC: -105.5 μM/kg FFM; P = .5842), rate of glucose disappearance was lower (difference in iAUC: 1494.2 μM/kg FFM; P = .0083). SI and liver insulin sensitivity were not different between study visits (P > .05). CONCLUSIONS Inhibition of glucagon and gastric emptying delaying reduced 2-hour prandial glucose excursions in T1D by delaying meal rate of glucose appearance.
Collapse
Affiliation(s)
- Ling Hinshaw
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| | - Michele Schiavon
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| | - Vikash Dadlani
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| | - Ashwini Mallad
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| | - Chiara Dalla Man
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| | - Adil Bharucha
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| | - Rita Basu
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| | - Jennifer R Geske
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| | - Rickey E Carter
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| | - Claudio Cobelli
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| | - Ananda Basu
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| | - Yogish C Kudva
- Division of Endocrinology and Metabolism (L.H., V.D., A.M., R.B., A.B., Y.C.K.), Mayo Clinic, Rochester, Minnesota; Department of Information Engineering (M.S., C.D.M., C.C.), University of Padova, Padova, Italy; Division of Gastroenterology (A.B.), Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research (J.R.G., R.E.C.), Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
153
|
Nutritional regulation of the anabolic fate of amino acids within the liver in mammals: concepts arising from in vivo studies. Nutr Res Rev 2016; 28:22-41. [PMID: 26156215 DOI: 10.1017/s0954422415000013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
At the crossroad between nutrient supply and requirements, the liver plays a central role in partitioning nitrogenous nutrients among tissues. The present review examines the utilisation of amino acids (AA) within the liver in various physiopathological states in mammals and how the fates of AA are regulated. AA uptake by the liver is generally driven by the net portal appearance of AA. This coordination is lost when demands by peripheral tissues is important (rapid growth or lactation), or when certain metabolic pathways within the liver become a priority (synthesis of acute-phase proteins). Data obtained in various species have shown that oxidation of AA and export protein synthesis usually responds to nutrient supply. Gluconeogenesis from AA is less dependent on hepatic delivery and the nature of nutrients supplied, and hormones like insulin are involved in the regulatory processes. Gluconeogenesis is regulated by nutritional factors very differently between mammals (glucose absorbed from the diet is important in single-stomached animals, while in carnivores, glucose from endogenous origin is key). The underlying mechanisms explaining how the liver adapts its AA utilisation to the body requirements are complex. The highly adaptable hepatic metabolism must be capable to deal with the various nutritional/physiological challenges that mammals have to face to maintain homeostasis. Whereas the liver responds generally to nutritional parameters in various physiological states occurring throughout life, other complex signalling pathways at systemic and tissue level (hormones, cytokines, nutrients, etc.) are involved additionally in specific physiological/nutritional states to prioritise certain metabolic pathways (pathological states or when nutritional requirements are uncovered).
Collapse
|
154
|
Hill MJ, Suzuki S, Segars JH, Kino T. CRTC2 Is a Coactivator of GR and Couples GR and CREB in the Regulation of Hepatic Gluconeogenesis. Mol Endocrinol 2016; 30:104-17. [PMID: 26652733 PMCID: PMC4695631 DOI: 10.1210/me.2015-1237] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/25/2015] [Indexed: 12/22/2022] Open
Abstract
Glucocorticoid hormones play essential roles in the regulation of gluconeogenesis in the liver, an adaptive response that is required for the maintenance of circulating glucose levels during fasting. Glucocorticoids do this by cooperating with glucagon, which is secreted from pancreatic islets to activate the cAMP-signaling pathway in hepatocytes. The cAMP-response element-binding protein (CREB)-regulated transcription coactivator 2 (CRTC2) is a coactivator known to be specific to CREB and plays a central role in the glucagon-mediated activation of gluconeogenesis in the early phase of fasting. We show here that CRTC2 also functions as a coactivator for the glucocorticoid receptor (GR). CRTC2 strongly enhances GR-induced transcriptional activity of glucocorticoid-responsive genes. CRTC2 physically interacts with the ligand-binding domain of the GR through a region spanning amino acids 561-693. Further, CRTC2 is required for the glucocorticoid-associated cooperative mRNA expression of the glucose-6-phosphatase, a rate-limiting enzyme for hepatic gluconeogenesis, by facilitating the attraction of GR and itself to its promoter region already occupied by CREB. CRTC2 is required for the maintenance of blood glucose levels during fasting in mice by enhancing the GR transcriptional activity on both the G6p and phosphoenolpyruvate carboxykinase (Pepck) genes. Finally, CRTC2 modulates the transcriptional activity of the progesterone receptor, indicating that it may influence the transcriptional activity of other steroid/nuclear receptors. Taken together, these results reveal that CRTC2 plays an essential role in the regulation of hepatic gluconeogenesis through coordinated regulation of the glucocorticoid/GR- and glucagon/CREB-signaling pathways on the key genes G6P and PEPCK.
Collapse
Affiliation(s)
- Micah J Hill
- Program in Reproductive and Adult Endocrinology (M.J.H., S.S., J.H.S., T.K.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; Division of Reproductive Endocrinology and Infertility (M.J.H.), Walter Reed National Military Medical Center, Bethesda, Maryland 20889; Department of Pediatrics (S.S.), Asahikawa Medical University, Asahikawa 078-8510, Japan; Division of Reproductive Sciences and Women's Health Research (J.H.S.), Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205; and Department of Experimental Therapeutics (T.K.), Division of Experimental Biology, Sidra Medical and Research Center, Doha 26999, Qatar
| | - Shigeru Suzuki
- Program in Reproductive and Adult Endocrinology (M.J.H., S.S., J.H.S., T.K.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; Division of Reproductive Endocrinology and Infertility (M.J.H.), Walter Reed National Military Medical Center, Bethesda, Maryland 20889; Department of Pediatrics (S.S.), Asahikawa Medical University, Asahikawa 078-8510, Japan; Division of Reproductive Sciences and Women's Health Research (J.H.S.), Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205; and Department of Experimental Therapeutics (T.K.), Division of Experimental Biology, Sidra Medical and Research Center, Doha 26999, Qatar
| | - James H Segars
- Program in Reproductive and Adult Endocrinology (M.J.H., S.S., J.H.S., T.K.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; Division of Reproductive Endocrinology and Infertility (M.J.H.), Walter Reed National Military Medical Center, Bethesda, Maryland 20889; Department of Pediatrics (S.S.), Asahikawa Medical University, Asahikawa 078-8510, Japan; Division of Reproductive Sciences and Women's Health Research (J.H.S.), Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205; and Department of Experimental Therapeutics (T.K.), Division of Experimental Biology, Sidra Medical and Research Center, Doha 26999, Qatar
| | - Tomoshige Kino
- Program in Reproductive and Adult Endocrinology (M.J.H., S.S., J.H.S., T.K.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; Division of Reproductive Endocrinology and Infertility (M.J.H.), Walter Reed National Military Medical Center, Bethesda, Maryland 20889; Department of Pediatrics (S.S.), Asahikawa Medical University, Asahikawa 078-8510, Japan; Division of Reproductive Sciences and Women's Health Research (J.H.S.), Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205; and Department of Experimental Therapeutics (T.K.), Division of Experimental Biology, Sidra Medical and Research Center, Doha 26999, Qatar
| |
Collapse
|
155
|
Affiliation(s)
- Ananda Basu
- Endocrine Research Unit, Division of Endocrinology and Metabolism, Saint Mary's Hospital , Mayo Clinic, Rochester, Minnesota
| | - Rita Basu
- Endocrine Research Unit, Division of Endocrinology and Metabolism, Saint Mary's Hospital , Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
156
|
Tsai JJ, Kuo HC, Lee KF, Tsai TH. Proteomic analysis of plasma from rats following total parenteral nutrition-induced liver injury. Proteomics 2015; 15:3865-74. [DOI: 10.1002/pmic.201500128] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/22/2015] [Accepted: 08/24/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Jai-Jen Tsai
- Division of Gastroenterology; Department of Medicine; National Yang-Ming University Hospital; I-Lan Taiwan
- Institute of Traditional Medicine; School of Medicine; National Yang-Ming University; Taipei Taiwan
| | - Hsing-Chun Kuo
- Department of Nursing; Chang Gung University of Science and Technology, Chronic Diseases and Health Promotion Research Center CGUST; Taiwan
- Research Center for Industry of Human Ecology; Chang Gung University of Science and Technology; Taoyuan Taiwan
| | - Kam-Fai Lee
- Department of Pathology; Chang Gung Memorial Hospital at Chiayi; Taiwan
| | - Tung-Hu Tsai
- Institute of Traditional Medicine; School of Medicine; National Yang-Ming University; Taipei Taiwan
- School of Pharmacy; College of Pharmacy; Kaohsiung Medical University; Kaohsiung Taiwan
- Department of Education and Research; Taipei City Hospital; Taipei Taiwan
| |
Collapse
|
157
|
Nixon M, Stewart-Fitzgibbon R, Fu J, Akhmedov D, Rajendran K, Mendoza-Rodriguez MG, Rivera-Molina YA, Gibson M, Berglund ED, Justice NJ, Berdeaux R. Skeletal muscle salt inducible kinase 1 promotes insulin resistance in obesity. Mol Metab 2015; 5:34-46. [PMID: 26844205 PMCID: PMC4703802 DOI: 10.1016/j.molmet.2015.10.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 10/19/2015] [Accepted: 10/22/2015] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE Insulin resistance causes type 2 diabetes mellitus and hyperglycemia due to excessive hepatic glucose production and inadequate peripheral glucose uptake. Our objectives were to test the hypothesis that the proposed CREB/CRTC2 inhibitor salt inducible kinase 1 (SIK1) contributes to whole body glucose homeostasis in vivo by regulating hepatic transcription of gluconeogenic genes and also to identify novel SIK1 actions on glucose metabolism. METHODS We created conditional (floxed) SIK1-knockout mice and studied glucose metabolism in animals with global, liver, adipose or skeletal muscle Sik1 deletion. We examined cAMP-dependent regulation of SIK1 and the consequences of SIK1 depletion on primary mouse hepatocytes. We probed metabolic phenotypes in tissue-specific SIK1 knockout mice fed high fat diet through hyperinsulinemic-euglycemic clamps and biochemical analysis of insulin signaling. RESULTS SIK1 knockout mice are viable and largely normoglycemic on chow diet. On high fat diet, global SIK1 knockout animals are strikingly protected from glucose intolerance, with both increased plasma insulin and enhanced peripheral insulin sensitivity. Surprisingly, liver SIK1 is not required for regulation of CRTC2 and gluconeogenesis, despite contributions of SIK1 to hepatocyte CRTC2 and gluconeogenesis regulation ex vivo. Sik1 mRNA accumulates in skeletal muscle of obese high fat diet-fed mice, and knockout of SIK1 in skeletal muscle, but not liver or adipose tissue, improves insulin sensitivity and muscle glucose uptake on high fat diet. CONCLUSIONS SIK1 is dispensable for glycemic control on chow diet. SIK1 promotes insulin resistance on high fat diet by a cell-autonomous mechanism in skeletal muscle. Our study establishes SIK1 as a promising therapeutic target to improve skeletal muscle insulin sensitivity in obese individuals without deleterious effects on hepatic glucose production.
Collapse
Key Words
- AKT, protein kinase B
- AMPK, AMP-activated protein kinase
- BAT, brown adipose tissue
- CHX, cycloheximide
- CREB
- CREB, cAMP response element-binding protein
- CRTC
- CRTC, CREB regulated transcription coactivator
- EndoRa, endogenous rate of glucose appearance
- FGF21, fibroblast growth factor 21
- FOXO1, forkhead box protein O1
- FSK, forskolin
- G6pase, glucose 6-phosphatase
- GDR, glucose disposal rate
- GIR, glucose infusion rate
- GTT, glucose tolerance test
- Glgn, glucagon
- Gluconeogenesis
- Glut, glucose transporter
- HDAC, histone deacetylase
- HFD, high fat diet
- HSP, heat shock protein
- IBMX, 3-isobutyl-1-methylxantine
- ITT, insulin tolerance test
- Insulin resistance
- PTT, pyruvate tolerance test
- Pepck, phosphoenolpyruvate carboxykinase
- Pgc, peroxisome proliferator-activated receptor gamma coactivator
- SIK, salt inducible kinase
- SIK1
- Salt inducible kinase
- WAT, white adipose tissue
- cAMP, cyclic adenosine monophosphate
Collapse
Affiliation(s)
- Mark Nixon
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Randi Stewart-Fitzgibbon
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA; Program in Cell and Regulatory Biology, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston TX 77030, USA
| | - Jingqi Fu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Dmitry Akhmedov
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Kavitha Rajendran
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Maria G Mendoza-Rodriguez
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Yisel A Rivera-Molina
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Micah Gibson
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Eric D Berglund
- Advanced Imaging Research Center and Department of Pharmacology, University of Texas Southwestern Medical School, USA
| | - Nicholas J Justice
- Institute of Molecular Medicine Center for Metabolic and Degenerative Diseases, University of Texas Health Science Center, Houston, TX 77030, USA; Program in Cell and Regulatory Biology, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston TX 77030, USA
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA; Institute of Molecular Medicine Center for Metabolic and Degenerative Diseases, University of Texas Health Science Center, Houston, TX 77030, USA; Program in Cell and Regulatory Biology, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston TX 77030, USA
| |
Collapse
|
158
|
Seoane-Collazo P, Fernø J, Gonzalez F, Diéguez C, Leis R, Nogueiras R, López M. Hypothalamic-autonomic control of energy homeostasis. Endocrine 2015; 50:276-91. [PMID: 26089260 DOI: 10.1007/s12020-015-0658-y] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 06/06/2015] [Indexed: 10/23/2022]
Abstract
Regulation of energy homeostasis is tightly controlled by the central nervous system (CNS). Several key areas such as the hypothalamus and brainstem receive and integrate signals conveying energy status from the periphery, such as leptin, thyroid hormones, and insulin, ultimately leading to modulation of food intake, energy expenditure (EE), and peripheral metabolism. The autonomic nervous system (ANS) plays a key role in the response to such signals, innervating peripheral metabolic tissues, including brown and white adipose tissue (BAT and WAT), liver, pancreas, and skeletal muscle. The ANS consists of two parts, the sympathetic and parasympathetic nervous systems (SNS and PSNS). The SNS regulates BAT thermogenesis and EE, controlled by central areas such as the preoptic area (POA) and the ventromedial, dorsomedial, and arcuate hypothalamic nuclei (VMH, DMH, and ARC). The SNS also regulates lipid metabolism in WAT, controlled by the lateral hypothalamic area (LHA), VMH, and ARC. Control of hepatic glucose production and pancreatic insulin secretion also involves the LHA, VMH, and ARC as well as the dorsal vagal complex (DVC), via splanchnic sympathetic and the vagal parasympathetic nerves. Muscle glucose uptake is also controlled by the SNS via hypothalamic nuclei such as the VMH. There is recent evidence of novel pathways connecting the CNS and ANS. These include the hypothalamic AMP-activated protein kinase-SNS-BAT axis which has been demonstrated to be a key modulator of thermogenesis. In this review, we summarize current knowledge of the role of the ANS in the modulation of energy balance.
Collapse
Affiliation(s)
- Patricia Seoane-Collazo
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain.
| | - Johan Fernø
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
- Department of Clinical Science, K. G. Jebsen Center for Diabetes Research, University of Bergen, 5021, Bergen, Norway
| | - Francisco Gonzalez
- Department of Surgery, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
- Service of Ophthalmology, Complejo Hospitalario Universitario de Santiago de Compostela, 15706, Santiago de Compostela, Spain
| | - Carlos Diéguez
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Rosaura Leis
- Unit of Investigation in Nutrition, Growth and Human Development of Galicia, Pediatric Department (USC), Complexo Hospitalario Universitario de Santiago (IDIS/SERGAS), Santiago de Compostela, Spain
| | - Rubén Nogueiras
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Miguel López
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain.
| |
Collapse
|
159
|
Arrojo e Drigo R, Ali Y, Diez J, Srinivasan DK, Berggren PO, Boehm BO. New insights into the architecture of the islet of Langerhans: a focused cross-species assessment. Diabetologia 2015. [PMID: 26215305 DOI: 10.1007/s00125-015-3699-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The human genome project and its search for factors underlying human diseases has fostered a major human research effort. Therefore, unsurprisingly, in recent years we have observed an increasing number of studies on human islet cells, including disease approaches focusing on type 1 and type 2 diabetes. Yet, the field of islet and diabetes research relies on the legacy of rodent-based investigations, which have proven difficult to translate to humans, particularly in type 1 diabetes. Whole islet physiology and pathology may differ between rodents and humans, and thus a comprehensive cross-species as well as species-specific view on islet research is much needed. In this review we summarise the current knowledge of interspecies islet cytoarchitecture, and discuss its potential impact on islet function and future perspectives in islet pathophysiology research.
Collapse
Affiliation(s)
- Rafael Arrojo e Drigo
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Yusuf Ali
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Juan Diez
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Dinesh Kumar Srinivasan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Per-Olof Berggren
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore.
- Imperial College London, London, UK.
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska University Hospital L1, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Bernhard O Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore.
- Imperial College London, London, UK.
- Department of Internal Medicine 1, Ulm University Medical Centre, Ulm, Germany.
| |
Collapse
|
160
|
Montefusco F, Pedersen MG. Mathematical modelling of local calcium and regulated exocytosis during inhibition and stimulation of glucagon secretion from pancreatic alpha-cells. J Physiol 2015; 593:4519-30. [PMID: 26236035 DOI: 10.1113/jp270777] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/28/2015] [Indexed: 02/06/2023] Open
Abstract
Glucagon secretion from pancreatic alpha-cells is dysregulated in diabetes. Despite decades of investigations of the control of glucagon release by glucose and hormones, the underlying mechanisms are still debated. Recently, mathematical models have been applied to investigate the modification of electrical activity in alpha-cells as a result of glucose application. However, recent studies have shown that paracrine effects such as inhibition of glucagon secretion by glucagon-like peptide 1 (GLP-1) or stimulation of release by adrenaline involve cAMP-mediated effects downstream of electrical activity. In particular, depending of the intracellular cAMP concentration, specific types of Ca(2+) channels are inhibited or activated, which interacts with mobilization of secretory granules. To investigate these aspects of alpha-cell function theoretically, we carefully developed a mathematical model of Ca(2+) levels near open or closed Ca(2+) channels of various types, which was linked to a description of Ca(2+) below the plasma membrane, in the bulk cytosol and in the endoplasmic reticulum. We investigated how the various subcellular Ca(2+) compartments contribute to control of glucagon-exocytosis in response to glucose, GLP-1 or adrenaline. Our studies refine previous modelling studies of alpha-cell function, and provide deeper insight into the control of glucagon secretion.
Collapse
Affiliation(s)
- Francesco Montefusco
- Department of Information Engineering, University of Padova, Via Gradenigo 6/B, 35131, Padova, Italy
| | - Morten Gram Pedersen
- Department of Information Engineering, University of Padova, Via Gradenigo 6/B, 35131, Padova, Italy
| |
Collapse
|
161
|
Pacini G, Ahrén B. Glucagon and GLP-1 exhibit no synergistic enhancement of glucose-stimulated insulin secretion in mice. Peptides 2015; 71:66-71. [PMID: 26119909 DOI: 10.1016/j.peptides.2015.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 05/25/2015] [Indexed: 11/25/2022]
Abstract
The combination of glucagon and glucagon-like peptide-1 (GLP-1) has been suggested as an approach to target obesity, since the two hormones have complementary action on body weight. We examined whether complementary action of the two hormones also exist on insulin secretion. Female C57BL/6 mice were injected intravenously with glucose with or without GLP-1, glucagon or the combination of GLP-1 and glucagon at three different dose levels. Furthermore, freshly isolated mouse islets were incubated for 30min in the presence of 2.8, 11.1 or 16.7mmol/l glucose or with 11.1mmol/l glucose in the presence of 100nmol/l glucagon and/or GLP-1. It was found that at 1min after glucose injection alone, insulin rose to a peak level and this peak, as well as the 50min area under the insulin curve (AUC insulin) were dose-dependently augmented by GLP-1 and glucagon. However, peak insulin with the two hormones together (with glucose) was not higher than after either single administration at any of the tested doses, i.e., no additive of synergistic action was observed by the combination on glucose-stimulated insulin secretion. Similar results were observed when calculating insulin for the whole test period. Also in vitro, both glucagon and GLP-1 augmented insulin secretion; however, there was no difference between the combined stimulation of insulin secretion by GLP-1 and glucagon together compared with either hormone alone. Insulin sensitivity did not exhibit significant changes from the glucose only condition. We conclude that the acute combined administration of the strongly insulinotropic GLP-1 and glucagon, both in vivo and in vitro, did not induce any additive or synergistic action on glucose-stimulated insulin secretion. This shows that the risk of a marked insulinotropic action when the two compounds are given together most likely does not result in increased risk of hypoglycemia.
Collapse
Affiliation(s)
- Giovanni Pacini
- Metabolic Unit, Institute of Neurosciences (IN-CNR), Corso Stati Uniti 4, 35127 Padova, Italy.
| | - Bo Ahrén
- Biomedical Center, C11, Department of Clinical Sciences, Lund University, SE 22184 Lund, Sweden
| |
Collapse
|
162
|
Hinshaw L, Mallad A, Dalla Man C, Basu R, Cobelli C, Carter RE, Kudva YC, Basu A. Glucagon sensitivity and clearance in type 1 diabetes: insights from in vivo and in silico experiments. Am J Physiol Endocrinol Metab 2015; 309:E474-86. [PMID: 26152766 PMCID: PMC4556882 DOI: 10.1152/ajpendo.00236.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/29/2015] [Indexed: 11/22/2022]
Abstract
Glucagon use in artificial pancreas for type 1 diabetes (T1D) is being explored for prevention and rescue from hypoglycemia. However, the relationship between glucagon stimulation of endogenous glucose production (EGP) viz., hepatic glucagon sensitivity, and prevailing glucose concentrations has not been examined. To test the hypothesis that glucagon sensitivity is increased at hypoglycemia vs. euglycemia, we studied 29 subjects with T1D randomized to a hypoglycemia or euglycemia clamp. Each subject was studied at three glucagon doses at euglycemia or hypoglycemia, with EGP measured by isotope dilution technique. The peak EGP increments and the integrated EGP response increased with increasing glucagon dose during euglycemia and hypoglycemia. However, the difference in dose response based on glycemia was not significant despite higher catecholamine concentrations in the hypoglycemia group. Knowledge of glucagon's effects on EGP was used to develop an in silico glucagon action model. The model-derived output fitted the obtained data at both euglycemia and hypoglycemia for all glucagon doses tested. Glucagon clearance did not differ between glucagon doses studied in both groups. Therefore, the glucagon controller of a dual hormone control system may not need to adjust glucagon sensitivity, and hence glucagon dosing, based on glucose concentrations during euglycemia and hypoglycemia.
Collapse
Affiliation(s)
- Ling Hinshaw
- Endocrine Research Unit, Division of Endocrinology, Mayo College of Medicine, Rochester, Minnesota
| | - Ashwini Mallad
- Endocrine Research Unit, Division of Endocrinology, Mayo College of Medicine, Rochester, Minnesota
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padua, Italy
| | - Rita Basu
- Endocrine Research Unit, Division of Endocrinology, Mayo College of Medicine, Rochester, Minnesota;
| | - Claudio Cobelli
- Department of Information Engineering, University of Padova, Padua, Italy
| | - Rickey E Carter
- Department of Health Sciences Research, Mayo College of Medicine, Rochester, Minnesota; and
| | - Yogish C Kudva
- Endocrine Research Unit, Division of Endocrinology, Mayo College of Medicine, Rochester, Minnesota
| | - Ananda Basu
- Endocrine Research Unit, Division of Endocrinology, Mayo College of Medicine, Rochester, Minnesota
| |
Collapse
|
163
|
Urocortin3 mediates somatostatin-dependent negative feedback control of insulin secretion. Nat Med 2015; 21:769-76. [PMID: 26076035 PMCID: PMC4496282 DOI: 10.1038/nm.3872] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/30/2015] [Indexed: 12/15/2022]
Abstract
The peptide hormone urocortin3 (Ucn3) is abundantly expressed by mature beta cells, yet its physiological role is unknown. Here we demonstrate that Ucn3 is stored and co-released with insulin and potentiates glucose-stimulated somatostatin secretion via cognate receptors on delta cells. Further, we found that islets lacking endogenous Ucn3 have fewer delta cells, reduced somatostatin content, impaired somatostatin secretion, and exaggerated insulin release, and that these defects are rectified by treatment with synthetic Ucn3 in vitro. Our observations indicate that the paracrine actions of Ucn3 activate a negative feedback loop that promotes somatostatin release to ensure the timely reduction of insulin secretion upon normalization of plasma glucose. Moreover, Ucn3 is markedly depleted from beta cells in mouse and macaque models of diabetes and in human diabetic islets. This suggests that Ucn3 is a key contributor to stable glycemic control, whose reduction during diabetes aggravates glycemic volatility and contributes to the pathophysiology of this disease.
Collapse
|
164
|
Sandoval DA, D'Alessio DA. Physiology of proglucagon peptides: role of glucagon and GLP-1 in health and disease. Physiol Rev 2015; 95:513-48. [PMID: 25834231 DOI: 10.1152/physrev.00013.2014] [Citation(s) in RCA: 330] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The preproglucagon gene (Gcg) is expressed by specific enteroendocrine cells (L-cells) of the intestinal mucosa, pancreatic islet α-cells, and a discrete set of neurons within the nucleus of the solitary tract. Gcg encodes multiple peptides including glucagon, glucagon-like peptide-1, glucagon-like peptide-2, oxyntomodulin, and glicentin. Of these, glucagon and GLP-1 have received the most attention because of important roles in glucose metabolism, involvement in diabetes and other disorders, and application to therapeutics. The generally accepted model is that GLP-1 improves glucose homeostasis indirectly via stimulation of nutrient-induced insulin release and by reducing glucagon secretion. Yet the body of literature surrounding GLP-1 physiology reveals an incompletely understood and complex system that includes peripheral and central GLP-1 actions to regulate energy and glucose homeostasis. On the other hand, glucagon is established principally as a counterregulatory hormone, increasing in response to physiological challenges that threaten adequate blood glucose levels and driving glucose production to restore euglycemia. However, there also exists a potential role for glucagon in regulating energy expenditure that has recently been suggested in pharmacological studies. It is also becoming apparent that there is cross-talk between the proglucagon derived-peptides, e.g., GLP-1 inhibits glucagon secretion, and some additive or synergistic pharmacological interaction between GLP-1 and glucagon, e.g., dual glucagon/GLP-1 agonists cause more weight loss than single agonists. In this review, we discuss the physiological functions of both glucagon and GLP-1 by comparing and contrasting how these peptides function, variably in concert and opposition, to regulate glucose and energy homeostasis.
Collapse
Affiliation(s)
- Darleen A Sandoval
- Division of Endocrinology and Metabolism, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David A D'Alessio
- Division of Endocrinology and Metabolism, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
165
|
Abstract
Artificial pancreas (AP) systems, a long-sought quest to replicate mechanically islet physiology that is lost in diabetes, are reaching the clinic, and the potential of automating insulin delivery is about to be realized. Significant progress has been made, and the safety and feasibility of AP systems have been demonstrated in the clinical research center and more recently in outpatient "real-world" environments. An iterative road map to AP system development has guided AP research since 2009, but progress in the field indicates that it needs updating. While it is now clear that AP systems are technically feasible, it remains much less certain that they will be widely adopted by clinicians and patients. Ultimately, the true success of AP systems will be defined by successful integration into the diabetes health care system and by the ultimate metric: improved diabetes outcomes.
Collapse
|
166
|
Kokil GR, Veedu RN, Ramm GA, Prins JB, Parekh HS. Type 2 diabetes mellitus: limitations of conventional therapies and intervention with nucleic acid-based therapeutics. Chem Rev 2015; 115:4719-43. [PMID: 25918949 DOI: 10.1021/cr5002832] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ganesh R Kokil
- †School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Rakesh N Veedu
- §Center for Comparative Genomics, Murdoch University, 90 South Street, Murdoch, WA 6150, Australia.,∥Western Australian Neuroscience Research Institute, Perth, WA 6150, Australia.,‡School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane QLD 4072 Australia
| | - Grant A Ramm
- ⊥The Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia.,#Faculty of Medicine and Biomedical Sciences, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Johannes B Prins
- ∇Mater Research Institute, The University of Queensland, Brisbane, QLD 4101, Australia
| | - Harendra S Parekh
- †School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Brisbane, QLD 4102, Australia
| |
Collapse
|
167
|
Yabe D, Nishikino R, Kaneko M, Iwasaki M, Seino Y. Short-term impacts of sodium/glucose co-transporter 2 inhibitors in Japanese clinical practice: considerations for their appropriate use to avoid serious adverse events. Expert Opin Drug Saf 2015; 14:795-800. [PMID: 25851664 DOI: 10.1517/14740338.2015.1034105] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Sodium/glucose co-transporter 2 inhibitors (SGLT2i) represent a novel class of glucose-lowering agents that lower plasma glucose levels through pharmacological inhibition of glucose reuptake from the kidney, independent of insulin secretion and action. Clinical trials of SGLT2i demonstrated therapeutic benefits on glycemic control and bodyweight in individuals with type 2 diabetes, with few cases of serious adverse events (SAEs). However, a considerable number of SAEs were reported in patients receiving SGLT2i clinically in Japan during the first 3 months of their use. These included urogenital infections, hypoglycemia and dehydration. Unexpectedly, serious skin and subcutaneous disorders, mainly reported as generalized rash or skin eruption, were prominent in patients receiving SGLT2i, but with unknown mechanisms. There is also concern for potential SAEs associated with chronic SGLT2i administration, especially in the non-obese type 2 diabetes characterized by reduced insulin secretion often seen in East Asia. Chronic SAEs may include severe hypoglycemia due to depletion of hepatic glycogen storage, acceleration of diabetes-associated sarcopenia and ketosis/ketoacidosis. The current information on acute SAEs confirms the importance of caution in the appropriate use of SGLT2i. Furthermore, careful long-term observation of patients receiving SGLT2i is essential to avoid SAEs and for better clinical use of this drug class.
Collapse
Affiliation(s)
- Daisuke Yabe
- Kansai Electric Power Hospital, Center for Diabetes, Endocrinology and Metabolism , 2-1-7 Fukushima-ku, Osaka 553-0003 , Japan +81 6 6458 5821 ; +81 6 6458 6994 ; ;
| | | | | | | | | |
Collapse
|
168
|
van der Meulen T, Huising MO. Role of transcription factors in the transdifferentiation of pancreatic islet cells. J Mol Endocrinol 2015; 54:R103-17. [PMID: 25791577 PMCID: PMC4373662 DOI: 10.1530/jme-14-0290] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The α and β cells act in concert to maintain blood glucose. The α cells release glucagon in response to low levels of glucose to stimulate glycogenolysis in the liver. In contrast, β cells release insulin in response to elevated levels of glucose to stimulate peripheral glucose disposal. Despite these opposing roles in glucose homeostasis, α and β cells are derived from a common progenitor and share many proteins important for glucose sensing and hormone secretion. Results from recent work have underlined these similarities between the two cell types by revealing that β-to-α as well as α-to-β transdifferentiation can take place under certain experimental circumstances. These exciting findings highlight unexpected plasticity of adult islets and offer hope of novel therapeutic paths to replenish β cells in diabetes. In this review, we focus on the transcription factor networks that establish and maintain pancreatic endocrine cell identity and how they may be perturbed to facilitate transdifferentiation.
Collapse
Affiliation(s)
- Talitha van der Meulen
- Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA
| | - Mark O Huising
- Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA
| |
Collapse
|
169
|
Boido A, Ceriani V, Pontiroli AE. Glucagon for hypoglycemic episodes in insulin-treated diabetic patients: a systematic review and meta-analysis with a comparison of glucagon with dextrose and of different glucagon formulations. Acta Diabetol 2015; 52:405-12. [PMID: 25323325 DOI: 10.1007/s00592-014-0665-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/06/2014] [Indexed: 10/24/2022]
Abstract
AIMS Glucagon is used as an emergency drug in hypoglycemia, mainly when the patient is unconscious. A few studies report on ineffectiveness of glucagon in relieving hypoglycemia. The present systematic review and meta-analysis evaluate the effectiveness of glucagon alone and in comparison with dextrose and the effectiveness of intranasal glucagon in comparison with injected glucagon. METHODS Studies were grouped into three groups: (1) reports on glucagon ineffectiveness; (2) comparison of glucagon and dextrose; (3) comparison of intranasal glucagon and injected glucagon. In groups 2 and 3, only controlled studies were included in the analysis, whether randomized or non-randomized studies. Appropriate methodology (PRISMA statement) was adhered to, and publication bias was formally assessed. Sixteen studies, published in any language as full papers, were analysed to identify predictors of ineffectiveness, and they were included in a meta-analysis (random effects model) to study the effect of different strategies. Intervention effect (number of failures) was expressed as odds ratio (OR), with 95 % confidence intervals. RESULTS Failure rate ranged from 0.0 to 2.31 %, to 7.6 %, to 14.4 %, and to 59 %. Comparing glucagon and dextrose, the OR was 0.53 (0.20-1.42); comparing intranasal and intramuscular glucagon, the OR was 1.40 (0.18-10.93). Heterogeneity was low and not statistically significant. Publication bias was absent. CONCLUSIONS These data indicate that ineffectiveness of glucagon is unfrequent, not different from dextrose; in addition, intranasal and injected glucagon are similarly effective. In the case of failure, a second dose can be administered.
Collapse
Affiliation(s)
- Augusto Boido
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milano, Italy
| | | | | |
Collapse
|
170
|
Basso AMM, Pelegrini PB, Mulinari F, Costa MC, Viana AB, Silva LP, Grossi-de-Sa MF. Recombinant glucagon: a differential biological activity. AMB Express 2015; 5:20. [PMID: 25852997 PMCID: PMC4385203 DOI: 10.1186/s13568-015-0099-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/22/2015] [Indexed: 11/13/2022] Open
Abstract
In Brazil, there is a growing demand for specialised pharmaceuticals, and the high cost of their importation results in increasing costs, reaching US$ 1.34 billion in 2012 and US$ 1.61 billion in 2013. Worldwide expenses related to drugs could reach US$ 1.3 trillion in 2018, especially due to new treatments for hepatitis C and cancer. Specialised or high-cost pharmaceutical drugs used for the treatment of viral hepatitis, multiple sclerosis, HIV and diabetes are distributed free of charge by the Brazilian government. The glucagon peptide was included in this group of high-cost biopharmaceuticals in 2008. Although its main application is the treatment of hypoglycaemia in diabetic patients, it can also be used with patients in an alcoholic coma, for those patients with biliary tract pain, and as a bronchodilator. Therefore, in order to reduce biopharmaceutical production costs, the Brazilian government passed laws focusing on the development and increase of a National Pharmaceutical Industrial Centre, including the demand for the national production of glucagon. For that reason and given the importance and high cost of recombinant glucagon, the purpose of this study was to develop methods to improve production, purification and performance of the biological activity of recombinant glucagon. Glucagon was recombined into a plasmid vector containing a Glutathione S-transferase tag, and the peptide was expressed in a heterologous Escherichia coli system. After purification procedures and molecular analyses, the biological activity of this recombinant glucagon was examined using in vivo assays and showed a highly significant (p < 0.00001) and prolonged effect on glucose levels when compared with the standard glucagon. The experimental procedure described here facilitates the high level production of recombinant glucagon with an extended biological activity.
Collapse
Affiliation(s)
- Angelina M M Basso
- />Department of Molecular Pathology, University of Brasilia, Brasilia, DF Brazil
- />Laboratory of Plant-Pest Interaction, Embrapa – Genetic Resources and Biotechnology, Brasília, DF Brazil
| | - Patrícia B Pelegrini
- />Laboratory of Plant-Pest Interaction, Embrapa – Genetic Resources and Biotechnology, Brasília, DF Brazil
| | - Fernanda Mulinari
- />Laboratory of Plant-Pest Interaction, Embrapa – Genetic Resources and Biotechnology, Brasília, DF Brazil
- />Pioneer Union for Social Insertion – UPIS, Planaltina, DF Brazil
| | - Michelle C Costa
- />Laboratory of Plant-Pest Interaction, Embrapa – Genetic Resources and Biotechnology, Brasília, DF Brazil
| | - Antonio B Viana
- />Laboratory of Plant-Pest Interaction, Embrapa – Genetic Resources and Biotechnology, Brasília, DF Brazil
- />Catholic University of Brasilia, Brasilia, DF Brazil
| | - Luciano P Silva
- />Laboratory of Mass Spectrometry, Embrapa – Genetic Resources and Biotechnology, Brasília, DF Brazil
| | - Maria Fatima Grossi-de-Sa
- />Laboratory of Plant-Pest Interaction, Embrapa – Genetic Resources and Biotechnology, Brasília, DF Brazil
- />Catholic University of Brasilia, Brasilia, DF Brazil
| |
Collapse
|
171
|
de Sousa MV, Fukui R, Krustrup P, Pereira RMR, Silva PRS, Rodrigues AC, de Andrade JL, Hernandez AJ, da Silva MER. Positive effects of football on fitness, lipid profile, and insulin resistance in Brazilian patients with type 2 diabetes. Scand J Med Sci Sports 2015; 24 Suppl 1:57-65. [PMID: 24944132 DOI: 10.1111/sms.12258] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2014] [Indexed: 12/19/2022]
Abstract
We evaluated the effects of recreational football training combined with calorie-restricted diet (football + diet) vs calorie-restricted diet alone (diet) on aerobic fitness, lipid profile, and insulin resistance indicators in type 2 diabetes (T2D) patients. Forty-four T2D patients aged 48-68 years (27 females, 17 males) were randomly allocated to the football + diet group (FDG; n = 22) or to the diet group (DG; n = 22), of whom 19 FDG and 15 DG subjects completed the study. The football training was performed for 3 × 40 min/week for 12 weeks. Dual-energy X-ray absorptiometry scanning, treadmill testing, and fasting blood samplings were performed pre and post-intervention. After 12 weeks, maximal oxygen uptake (VO₂max ) was elevated (P < 0.05) by 10 ± 4% in FDG but not in DG (-3 ± 4%, P < 0.05). After 12 weeks, reductions in blood triglycerides (0.4 ± 0.1 mmol/L), total cholesterol (0.6 ± 0.2 mmol/L), low-density lipoprotein, and very low-density lipoprotein levels were observed only in FDG. Fat mass decreased (P < 0.05) by 3.4 ± 0.4 kg in FDG and 3.7 ± 0.4 kg in DG. The lower (P < 0.05) glucagon and homeostatic model assessment of insulin resistance indicated an improvement in insulin sensitivity in FDG. In conclusion, football combined with restricted diet was effective in enhancing VO₂max , reducing total cholesterol and triglycerides, and increasing insulin sensitivity, potentially providing better tools for the prevention of T2D complications than diet alone.
Collapse
Affiliation(s)
- M V de Sousa
- Laboratory of Medical Investigation, LIM-18, Endocrinology Division, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Brun PJ, Grijalva A, Rausch R, Watson E, Yuen JJ, Das BC, Shudo K, Kagechika H, Leibel RL, Blaner WS. Retinoic acid receptor signaling is required to maintain glucose-stimulated insulin secretion and β-cell mass. FASEB J 2015; 29. [PMID: 25389133 PMCID: PMC4314234 DOI: 10.1096/fj.14-256743 10.1096/fj.14-256743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Retinoic acid signaling is required for maintaining a range of cellular processes, including cell differentiation, proliferation, and apoptosis. We investigated the actions of all-trans-retinoic acid (atRA) signaling in pancreatic β-cells of adult mice. atRA signaling was ablated in β-cells by overexpressing a dominant-negative retinoic acid receptor (RAR)-α mutant (RARdn) using an inducible Cre-Lox system under the control of the pancreas duodenal homeobox gene promoter. Our studies establish that hypomorphism for RAR in β-cells leads to an age-dependent decrease in plasma insulin in the fed state and in response to a glucose challenge. Glucose-stimulated insulin secretion was also impaired in islets isolated from mice expressing RARdn. Among genes that are atRA responsive, Glut2 and Gck mRNA levels were decreased in isolated islets from RARdn-expressing mice. Histologic analyses of RARdn-expressing pancreata revealed a decrease in β-cell mass and insulin per β-cell 1 mo after induction of the RARdn. Our results indicate that atRA signaling mediated by RARs is required in the adult pancreas for maintaining both β-cell function and mass, and provide insights into molecular mechanisms underlying these actions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - William S. Blaner
- Correspondence: Department of Medicine, College of Physicians and Surgeons, Columbia University, 630 W. 168th Street, New York, NY 10032, USA. E-mail:
| |
Collapse
|
173
|
Irwin DM. Evolution of receptors for peptides similar to glucagon. Gen Comp Endocrinol 2014; 209:50-60. [PMID: 24650782 DOI: 10.1016/j.ygcen.2014.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 02/24/2014] [Accepted: 03/05/2014] [Indexed: 12/25/2022]
Abstract
The genes encoding the peptide precursors for glucagon (GCG), glucose-dependent insulinotropic peptide (GIP), and ortholog of exendin belong to the same family as shown by sequence similarity. The peptides similar to glucagon encoded by these genes signal through a closely related subfamily of G-protein coupled receptors. A total of five types of genes for receptors for these peptides have been identified, three for the products of GCG (GCGR, GLP1R, and GLP2R) and one each for the products of GIP (GIPR) and the ortholog of exendin (Grlr). Phylogenetic and genomic neighborhood analyses clearly show that these genes originated very early in vertebrate evolution and all were present in the common ancestor of tetrapods and bony fish. Despite their ancient origins, some of these genes are dispensable, with the Glp1r, Gipr, and Grlr being lost on the lineages leading to bony fish, birds, and mammals, respectively. The loss of the genes for these receptors may have been driving forces in the evolution of new functions for these peptides similar to glucagon.
Collapse
Affiliation(s)
- David M Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ont. M5S 1A8, Canada; Banting and Best Diabetes Centre, University of Toronto, Toronto, Ont. M5S 1A8, Canada.
| |
Collapse
|
174
|
Brun PJ, Grijalva A, Rausch R, Watson E, Yuen JJ, Das BC, Shudo K, Kagechika H, Leibel RL, Blaner WS. Retinoic acid receptor signaling is required to maintain glucose-stimulated insulin secretion and β-cell mass. FASEB J 2014; 29:671-83. [PMID: 25389133 DOI: 10.1096/fj.14-256743] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Retinoic acid signaling is required for maintaining a range of cellular processes, including cell differentiation, proliferation, and apoptosis. We investigated the actions of all-trans-retinoic acid (atRA) signaling in pancreatic β-cells of adult mice. atRA signaling was ablated in β-cells by overexpressing a dominant-negative retinoic acid receptor (RAR)-α mutant (RARdn) using an inducible Cre-Lox system under the control of the pancreas duodenal homeobox gene promoter. Our studies establish that hypomorphism for RAR in β-cells leads to an age-dependent decrease in plasma insulin in the fed state and in response to a glucose challenge. Glucose-stimulated insulin secretion was also impaired in islets isolated from mice expressing RARdn. Among genes that are atRA responsive, Glut2 and Gck mRNA levels were decreased in isolated islets from RARdn-expressing mice. Histologic analyses of RARdn-expressing pancreata revealed a decrease in β-cell mass and insulin per β-cell 1 mo after induction of the RARdn. Our results indicate that atRA signaling mediated by RARs is required in the adult pancreas for maintaining both β-cell function and mass, and provide insights into molecular mechanisms underlying these actions.
Collapse
Affiliation(s)
- Pierre-Jacques Brun
- Departments of *Medicine and Pediatrics and Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY USA; Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, USA; Research Foundation Itsuu Laboratory, Tokyo, Japan; and Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ambar Grijalva
- Departments of *Medicine and Pediatrics and Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY USA; Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, USA; Research Foundation Itsuu Laboratory, Tokyo, Japan; and Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Richard Rausch
- Departments of *Medicine and Pediatrics and Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY USA; Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, USA; Research Foundation Itsuu Laboratory, Tokyo, Japan; and Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Elizabeth Watson
- Departments of *Medicine and Pediatrics and Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY USA; Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, USA; Research Foundation Itsuu Laboratory, Tokyo, Japan; and Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Jason J Yuen
- Departments of *Medicine and Pediatrics and Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY USA; Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, USA; Research Foundation Itsuu Laboratory, Tokyo, Japan; and Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Bhaskar C Das
- Departments of *Medicine and Pediatrics and Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY USA; Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, USA; Research Foundation Itsuu Laboratory, Tokyo, Japan; and Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koichi Shudo
- Departments of *Medicine and Pediatrics and Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY USA; Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, USA; Research Foundation Itsuu Laboratory, Tokyo, Japan; and Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Kagechika
- Departments of *Medicine and Pediatrics and Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY USA; Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, USA; Research Foundation Itsuu Laboratory, Tokyo, Japan; and Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Rudolph L Leibel
- Departments of *Medicine and Pediatrics and Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY USA; Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, USA; Research Foundation Itsuu Laboratory, Tokyo, Japan; and Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - William S Blaner
- Departments of *Medicine and Pediatrics and Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY USA; Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, USA; Research Foundation Itsuu Laboratory, Tokyo, Japan; and Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
175
|
El Youssef J, Castle JR, Bakhtiani PA, Haidar A, Branigan DL, Breen M, Ward WK. Quantification of the glycemic response to microdoses of subcutaneous glucagon at varying insulin levels. Diabetes Care 2014; 37:3054-60. [PMID: 25139882 PMCID: PMC4207205 DOI: 10.2337/dc14-0803] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Glucagon delivery in closed-loop control of type 1 diabetes is effective in minimizing hypoglycemia. However, high insulin concentration lowers the hyperglycemic effect of glucagon, and small doses of glucagon in this setting are ineffective. There are no studies clearly defining the relationship between insulin levels, subcutaneous glucagon, and blood glucose. RESEARCH DESIGN AND METHODS Using a euglycemic clamp technique in 11 subjects with type 1 diabetes, we examined endogenous glucose production (EGP) of glucagon (25, 75, 125, and 175 μg) at three insulin infusion rates (0.016, 0.032, and 0.05 units/kg/h) in a randomized, crossover study. Infused 6,6-dideuterated glucose was measured every 10 min, and EGP was determined using a validated glucoregulatory model. Area under the curve (AUC) for glucose production was the primary outcome, estimated over 60 min. RESULTS At low insulin levels, EGP rose proportionately with glucagon dose, from 5 ± 68 to 112 ± 152 mg/kg (P = 0.038 linear trend), whereas at high levels, there was no increase in glucose output (19 ± 53 to 26 ± 38 mg/kg, P = NS). Peak glucagon serum levels and AUC correlated well with dose (r2 = 0.63, P < 0.001), as did insulin levels with insulin infusion rates (r2 = 0.59, P < 0.001). CONCLUSIONS EGP increases steeply with glucagon doses between 25 and 175 μg at lower insulin infusion rates. However, high insulin infusion rates prevent these doses of glucagon from significantly increasing glucose output and may reduce glucagon effectiveness in preventing hypoglycemia when used in the artificial pancreas.
Collapse
Affiliation(s)
| | | | | | - Ahmad Haidar
- Institut de Recherches Cliniques de Montréal, Montreal, Canada
| | | | | | - W Kenneth Ward
- Oregon Health & Science University, Portland, OR Legacy Health, Portland, OR
| |
Collapse
|
176
|
Yan S, Zhang Q, Zhong X, Tang J, Wang Y, Yu J, Zhou Y, Zhang J, Guo F, Liu Y, FitzGerald GA, Yu Y. I prostanoid receptor-mediated inflammatory pathway promotes hepatic gluconeogenesis through activation of PKA and inhibition of AKT. Diabetes 2014; 63:2911-23. [PMID: 24722246 DOI: 10.2337/db13-1893] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs), including acetylsalicylic acid (ASA), improve glucose metabolism in diabetic subjects, although the underlying mechanisms remain unclear. In this study, we observed dysregulated expression of cyclooxygenase-2, prostacyclin biosynthesis, and the I prostanoid receptor (IP) in the liver's response to diabetic stresses. High doses of ASA reduced hepatic prostaglandin generation and suppressed hepatic gluconeogenesis in mice during fasting, and the hypoglycemic effect of ASA could be restored by IP agonist treatment. IP deficiency inhibited starvation-induced hepatic gluconeogenesis, thus inhibiting the progression of diabetes, whereas hepatic overexpression of IP increased gluconeogenesis. IP deletion depressed cAMP-dependent CREB phosphorylation and elevated AKT phosphorylation by suppressing PI3K-γ/PKC-ζ-mediated TRB3 expression, which subsequently downregulated the gluconeogenic genes for glucose-6-phosphatase (G6Pase) and phosphoenol pyruvate carboxykinase 1 in hepatocytes. We therefore conclude that suppression of IP modulation of hepatic gluconeogenesis through the PKA/CREB and PI3K-γ/PKC-ζ/TRB3/AKT pathways contributes to the effects of NSAIDs in diabetes.
Collapse
Affiliation(s)
- Shuai Yan
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qianqian Zhang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaojing Zhong
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Juan Tang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuanyang Wang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Junjie Yu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Zhou
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jian Zhang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Liu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Garret A FitzGerald
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA
| | - Ying Yu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| |
Collapse
|
177
|
Lindén J, Lensu S, Pohjanvirta R. Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on hormones of energy balance in a TCDD-sensitive and a TCDD-resistant rat strain. Int J Mol Sci 2014; 15:13938-66. [PMID: 25119860 PMCID: PMC4159833 DOI: 10.3390/ijms150813938] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/24/2014] [Accepted: 07/29/2014] [Indexed: 01/16/2023] Open
Abstract
One of the hallmarks of the acute toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a drastically reduced feed intake by an unknown mechanism. To further elucidate this wasting syndrome, we followed the effects of a single large dose (100 μg/kg) of TCDD on the serum levels of several energy balance-influencing hormones, clinical chemistry variables, and hepatic aryl hydrocarbon receptor (AHR) expression in two rat strains that differ widely in their TCDD sensitivities, for up to 10 days. TCDD affected most of the analytes in sensitive Long-Evans rats, while there were few alterations in the resistant Han/Wistar strain. However, analyses of feed-restricted unexposed Long-Evans rats indicated several of the perturbations to be secondary to energy deficiency. Notable increases in ghrelin and glucagon occurred in TCDD-treated Long-Evans rats alone, which links these hormones to the wasting syndrome. The newly found energy balance regulators, insulin-like growth factor 1 and fibroblast growth factor 21 (FGF-21), appeared to function in concert in body weight loss-induced metabolic state, and FGF-21 was putatively linked to increased lipolysis induced by TCDD. Finally, we demonstrate a reverse set of changes in the AHR protein and mRNA response to TCDD and feed restriction, suggesting that AHR might function also as a physiological regulator, possibly involved in the maintenance of energy balance.
Collapse
Affiliation(s)
- Jere Lindén
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, FI-00014 Helsinki, Finland.
| | - Sanna Lensu
- Department of Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35, FI-40014 Jyväskylä, Finland.
| | - Raimo Pohjanvirta
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, FI-00014 Helsinki, Finland.
| |
Collapse
|
178
|
Benner C, van der Meulen T, Cacéres E, Tigyi K, Donaldson CJ, Huising MO. The transcriptional landscape of mouse beta cells compared to human beta cells reveals notable species differences in long non-coding RNA and protein-coding gene expression. BMC Genomics 2014; 15:620. [PMID: 25051960 PMCID: PMC4124169 DOI: 10.1186/1471-2164-15-620] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/10/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Insulin producing beta cell and glucagon producing alpha cells are colocalized in pancreatic islets in an arrangement that facilitates the coordinated release of the two principal hormones that regulate glucose homeostasis and prevent both hypoglycemia and diabetes. However, this intricate organization has also complicated the determination of the cellular source(s) of the expression of genes that are detected in the islet. This reflects a significant gap in our understanding of mouse islet physiology, which reduces the effectiveness by which mice model human islet disease. RESULTS To overcome this challenge, we generated a bitransgenic reporter mouse that faithfully labels all beta and alpha cells in mouse islets to enable FACS-based purification and the generation of comprehensive transcriptomes of both populations. This facilitates systematic comparison across thousands of genes between the two major endocrine cell types of the islets of Langerhans whose principal hormones are of cardinal importance for glucose homeostasis. Our data leveraged against similar data for human beta cells reveal a core common beta cell transcriptome of 9900+ genes. Against the backdrop of overall similar beta cell transcriptomes, we describe marked differences in the repertoire of receptors and long non-coding RNAs between mouse and human beta cells. CONCLUSIONS The comprehensive mouse alpha and beta cell transcriptomes complemented by the comparison of the global (dis)similarities between mouse and human beta cells represent invaluable resources to boost the accuracy by which rodent models offer guidance in finding cures for human diabetes.
Collapse
Affiliation(s)
- Christopher Benner
- />Razzavi Newman Integrated Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Talitha van der Meulen
- />Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Elena Cacéres
- />Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Kristof Tigyi
- />Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Cynthia J Donaldson
- />Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Mark O Huising
- />Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
- />Department of Neurobiology, Physiology & Behavior, University of California, One Shields Avenue, 180 Briggs Hall, Davis, CA 95616 USA
| |
Collapse
|
179
|
Ittner AA, Bertz J, Chan TYB, van Eersel J, Polly P, Ittner LM. The nucleotide exchange factor SIL1 is required for glucose-stimulated insulin secretion from mouse pancreatic beta cells in vivo. Diabetologia 2014; 57:1410-9. [PMID: 24733160 DOI: 10.1007/s00125-014-3230-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 03/13/2014] [Indexed: 01/06/2023]
Abstract
AIMS/HYPOTHESIS Regulation of insulin secretion along the secretory pathway is incompletely understood. We addressed the expression of SIL1, a nucleotide exchange factor for the endoplasmic reticulum (ER) chaperone glucose-regulated protein 78 kD (GRP78), in pancreatic beta cells and investigated whether or not SIL1 is involved in beta cell function. METHODS SIL1 expression was analysed by immunoblotting and immunofluorescence. Metabolic and islet variables, including glucose tolerance, beta cell mass, insulin secretion, islet ultrastructure, insulin content and levels of ER stress marker proteins, were addressed in Sil1 knockout (Sil1 (-/-)) mice. Insulin, proinsulin and C-peptide release was addressed in Sil1 (-/-) islets, and SIL1 overexpression or knockdown was explored in MIN6 cells in vitro. Models of type 1 diabetes and insulin resistance were induced in Sil1 (-/-) mice by administration of streptozotocin (STZ) and a high-fat diet (HFD), respectively. RESULTS We show that SIL1 is expressed in pancreatic beta cells and is required for islet insulin content, islet sizing, glucose tolerance and glucose-stimulated insulin secretion in vivo. Levels of pancreatic ER stress markers are increased in Sil1 (-/-) mice, and Sil1 (-/-) beta cell ER is ultrastructurally compromised. Isolated Sil1 (-/-) islets show lower proinsulin and insulin content and impaired glucose-stimulated insulin secretion. Modulation of SIL1 protein levels in MIN6 cells correlates with changes in insulin content and secreted insulin. Furthermore, Sil1 (-/-) mice are more susceptible to STZ-induced type 1 diabetes with increased apoptosis. Upon HFD feeding, Sil1 (-/-) mice show markedly lower insulin secretion and exacerbated glucose intolerance compared with control mice. Surprisingly, however, HFD-fed Sil1 (-/-) mice display pronounced islet hyperplasia with low amounts of insulin in total pancreas. CONCLUSIONS/INTERPRETATION These results reveal a novel role for the nucleotide exchange factor SIL1 in pancreatic beta cell function under physiological and disease conditions such as diabetes and the metabolic syndrome.
Collapse
Affiliation(s)
- Arne A Ittner
- School of Medical Sciences, University of New South Wales, Botany Street, Kensington, Sydney, 2052, NSW, Australia,
| | | | | | | | | | | |
Collapse
|
180
|
Cullen KS, Al-Oanzi ZH, O'Harte FPM, Agius L, Arden C. Glucagon induces translocation of glucokinase from the cytoplasm to the nucleus of hepatocytes by transfer between 6-phosphofructo 2-kinase/fructose 2,6-bisphosphatase-2 and the glucokinase regulatory protein. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1843:1123-34. [PMID: 24566088 PMCID: PMC4024195 DOI: 10.1016/j.bbamcr.2014.02.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/07/2014] [Accepted: 02/12/2014] [Indexed: 01/28/2023]
Abstract
Glucokinase activity is a major determinant of hepatic glucose metabolism and blood glucose homeostasis. Liver glucokinase activity is regulated acutely by adaptive translocation between the nucleus and the cytoplasm through binding and dissociation from its regulatory protein (GKRP) in the nucleus. Whilst the effect of glucose on this mechanism is well established, the role of hormones in regulating glucokinase location and its interaction with binding proteins remains unsettled. Here we show that treatment of rat hepatocytes with 25mM glucose caused decreased binding of glucokinase to GKRP, translocation from the nucleus and increased binding to 6-phosphofructo 2-kinase/fructose 2,6 bisphosphatase-2 (PFK2/FBPase2) in the cytoplasm. Glucagon caused dissociation of glucokinase from PFK2/FBPase2, concomitant with phosphorylation of PFK2/FBPase2 on Ser-32, uptake of glucokinase into the nucleus and increased interaction with GKRP. Two novel glucagon receptor antagonists attenuated the action of glucagon. This establishes an unequivocal role for hormonal control of glucokinase translocation. Given that glucagon excess contributes to the pathogenesis of diabetes, glucagon may play a role in the defect in glucokinase translocation and activity evident in animal models and human diabetes.
Collapse
Affiliation(s)
- Kirsty S Cullen
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Ziad H Al-Oanzi
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK; Department of Laboratory Medicine, Al-Jouf University, Sakaka, Saudi Arabia
| | - Finbarr P M O'Harte
- The Saad Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, UK
| | - Loranne Agius
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Catherine Arden
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
181
|
Aw DKL, Sinha RA, Xie SY, Yen PM. Differential AMPK phosphorylation by glucagon and metformin regulates insulin signaling in human hepatic cells. Biochem Biophys Res Commun 2014; 447:569-573. [PMID: 24735537 DOI: 10.1016/j.bbrc.2014.04.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 04/07/2014] [Indexed: 01/10/2023]
Abstract
Insulin and glucagon signaling in the liver are major contributors to glucose homeostasis. Patients with Type 1 and Type 2 diabetes have impaired glycemic control due, in part, to dysregulation of the opposing actions of these hormones. While hyperglucagonemia is a common feature in diabetes, its precise role in insulin resistance is not well understood. Recently, metformin, an AMPK activator, was shown to regulate hepatic glucose output via inhibition of glucagon-induced cAMP/PKA signaling; however, the mechanism for how PKA inhibition leads to AMPK activation in human hepatic cells is not known. Here we show that glucagon impairs insulin-mediated AKT phosphorylation in human hepatic cell line Huh7. This impairment of AKT activation by glucagon is due to PKA-mediated inhibition of AMPK via increased inhibitory phosphorylation of AMPK(Ser173) and reduced activating phosphorylation of AMPK(Thr172). In contrast, metformin decreases PKA activity, leading to decreased pAMPK(Ser173) and increased pAMPK(Thr172). These data support a novel mechanism involving PKA-dependent AMPK phosphorylation that provides new insight into how glucagon and metformin modulate hepatic insulin resistance.
Collapse
Affiliation(s)
- Darius Kang Lie Aw
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore
| | - Rohit A Sinha
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore
| | - Sherwin Ying Xie
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore
| | - Paul M Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore; Sarah W. Stedman Nutrition and Metabolism Center, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
182
|
Suckow AT, Polidori D, Yan W, Chon S, Ma JY, Leonard J, Briscoe CP. Alteration of the glucagon axis in GPR120 (FFAR4) knockout mice: a role for GPR120 in glucagon secretion. J Biol Chem 2014; 289:15751-63. [PMID: 24742677 DOI: 10.1074/jbc.m114.568683] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
GPR40 (FFAR1) and GPR120 (FFAR4) are G-protein-coupled receptors (GPCRs) that are activated by long chain fatty acids (LCFAs). GPR40 is expressed at high levels in islets and mediates the ability of LCFAs to potentiate glucose-stimulated insulin secretion (GSIS). GPR120 is expressed at high levels in colon, adipose, and pituitary, and at more modest levels in pancreatic islets. The role of GPR120 in islets has not been explored extensively. Here, we confirm that saturated (e.g. palmitic acid) and unsaturated (e.g. docosahexaenoic acid (DHA)) LCFAs engage GPR120 and demonstrate that palmitate- and DHA-potentiated glucagon secretion are greatly reduced in isolated GPR120 KO islets. Remarkably, LCFA potentiated glucagon secretion is similarly reduced in GPR40 KO islets. Compensatory changes in mRNA expression of GPR120 in GPR40 KO islets, and vice versa, do not explain that LCFA potentiated glucagon secretion seemingly involves both receptors. LCFA-potentiated GSIS remains intact in GPR120 KO islets. Consistent with previous reports, GPR120 KO mice are hyperglycemic and glucose intolerant; however, our KO mice display evidence of a hyperactive counter-regulatory response rather than insulin resistance during insulin tolerance tests. An arginine stimulation test and a glucagon challenge confirmed both increases in glucagon secretion and liver glucagon sensitivity in GPR120 KO mice relative to WT mice. Our findings demonstrate that GPR120 is a nutrient sensor that is activated endogenously by both saturated and unsaturated long chain fatty acids and that an altered glucagon axis likely contributes to the impaired glucose homeostasis observed in GPR120 KO mice.
Collapse
Affiliation(s)
- Arthur T Suckow
- From Janssen Research & Development, LLC, San Diego, California 92121
| | - David Polidori
- From Janssen Research & Development, LLC, San Diego, California 92121
| | - Wen Yan
- From Janssen Research & Development, LLC, San Diego, California 92121
| | - Suhyoun Chon
- From Janssen Research & Development, LLC, San Diego, California 92121
| | - Jing Ying Ma
- From Janssen Research & Development, LLC, San Diego, California 92121
| | - James Leonard
- From Janssen Research & Development, LLC, San Diego, California 92121
| | - Celia P Briscoe
- From Janssen Research & Development, LLC, San Diego, California 92121
| |
Collapse
|
183
|
Oosterveer MH, Schoonjans K. Hepatic glucose sensing and integrative pathways in the liver. Cell Mol Life Sci 2014; 71:1453-67. [PMID: 24196749 PMCID: PMC11114046 DOI: 10.1007/s00018-013-1505-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/17/2013] [Accepted: 10/18/2013] [Indexed: 12/21/2022]
Abstract
The hepatic glucose-sensing system is a functional network of enzymes and transcription factors that is critical for the maintenance of energy homeostasis and systemic glycemia. Here we review the recent literature on its components and metabolic actions. Glucokinase (GCK) is generally considered as the initial postprandial glucose-sensing component, which acts as the gatekeeper for hepatic glucose metabolism and provides metabolites that activate the transcription factor carbohydrate response element binding protein (ChREBP). Recently, liver receptor homolog 1 (LRH-1) has emerged as an upstream regulator of the central GCK-ChREBP axis, with a critical role in the integration of hepatic intermediary metabolism in response to glucose. Evidence is also accumulating that O-linked β-N-acetylglucosaminylation (O-GlcNAcylation) and acetylation can act as glucose-sensitive modifications that may contribute to hepatic glucose sensing by targeting regulatory proteins and the epigenome. Further elucidation of the components and functional roles of the hepatic glucose-sensing system may contribute to the future treatment of liver diseases associated with deregulated glucose sensors.
Collapse
Affiliation(s)
- Maaike H. Oosterveer
- Department of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Kristina Schoonjans
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
184
|
Drougard A, Duparc T, Brenachot X, Carneiro L, Gouazé A, Fournel A, Geurts L, Cadoudal T, Prats AC, Pénicaud L, Vieau D, Lesage J, Leloup C, Benani A, Cani PD, Valet P, Knauf C. Hypothalamic apelin/reactive oxygen species signaling controls hepatic glucose metabolism in the onset of diabetes. Antioxid Redox Signal 2014; 20:557-73. [PMID: 23879244 PMCID: PMC3901354 DOI: 10.1089/ars.2013.5182] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
AIMS We have previously demonstrated that central apelin is implicated in the control of peripheral glycemia, and its action depends on nutritional (fast versus fed) and physiological (normal versus diabetic) states. An intracerebroventricular (icv) injection of a high dose of apelin, similar to that observed in obese/diabetic mice, increase fasted glycemia, suggesting (i) that apelin contributes to the establishment of a diabetic state, and (ii) the existence of a hypothalamic to liver axis. Using pharmacological, genetic, and nutritional approaches, we aim at unraveling this system of regulation by identifying the hypothalamic molecular actors that trigger the apelin effect on liver glucose metabolism and glycemia. RESULTS We show that icv apelin injection stimulates liver glycogenolysis and gluconeogenesis via an over-activation of the sympathetic nervous system (SNS), leading to fasted hyperglycemia. The effect of central apelin on liver function is dependent of an increased production of hypothalamic reactive oxygen species (ROS). These data are strengthened by experiments using lentiviral vector-mediated over-expression of apelin in hypothalamus of mice that present over-activation of SNS associated to an increase in hepatic glucose production. Finally, we report that mice fed a high-fat diet present major alterations of hypothalamic apelin/ROS signaling, leading to activation of glycogenolysis. INNOVATION/CONCLUSION: These data bring compelling evidence that hypothalamic apelin is one master switch that participates in the onset of diabetes by directly acting on liver function. Our data support the idea that hypothalamic apelin is a new potential therapeutic target to treat diabetes.
Collapse
Affiliation(s)
- Anne Drougard
- 1 Institut National de la Santé et de la Recherche Médicale (INSERM) , Toulouse, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Bernsmeier C, Calabrese D, Heim MH, Duong HTF. Hepatitis C virus dysregulates glucose homeostasis by a dual mechanism involving induction of PGC1α and dephosphorylation of FoxO1. J Viral Hepat 2014; 21:9-18. [PMID: 24329853 DOI: 10.1111/jvh.12208] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 10/15/2013] [Indexed: 12/11/2022]
Abstract
The maintenance of glucose homeostasis is a complex process in which the insulin signalling pathway plays a major role. Disruption of insulin-regulated glucose homeostasis is frequently observed in chronic hepatitis C (CHC) infection and might potentially contribute to type 2 diabetes mellitus (T2DM) development. Presently, the mechanism that links HCV infection to insulin resistance remains unclear. Previously, we have reported that HCV protein expression in HCV transgenic mice (B6HCV) leads to an overexpression of protein phosphatase 2A (PP2A) through an ER stress response. In the present work, we describe an association of FoxO1 hypophosphorylation and upregulation of both PGC-1α and G6Pase to phenotypic hyperglycaemia and insulin resistance in B6HCV mice. In vitro, we observed that PGC1α is concomitantly induced with PP2A. Moreover, we show that the enhanced PP2A expression is sufficient to inhibit insulin-induced FoxO1 phosphorylation via blockade of insulin-mediated Akt activation or/and through direct association and dephosphorylation of pS-FoxO1. Consequently, we found that the gluconeogenic gene glucose-6-phosphatase is upregulated. These observations were confirmed in liver biopsies obtained from CHC patients. In summary, our results show that HCV-mediated upregulation of PP2A catalytic subunit alters signalling pathways that control hepatic glucose homeostasis by inhibiting Akt and dephosphorylation of FoxO1.
Collapse
Affiliation(s)
- C Bernsmeier
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland; Division of Gastroenterology and Hepatology, University of Basel, Basel, Switzerland
| | | | | | | |
Collapse
|
186
|
Xu X, Hu J, McGrath BC, Cavener DR. GCN2 regulates the CCAAT enhancer binding protein beta and hepatic gluconeogenesis. Am J Physiol Endocrinol Metab 2013; 305:E1007-17. [PMID: 23900421 PMCID: PMC3798698 DOI: 10.1152/ajpendo.00063.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mice deficient for general control nondepressible-2 (Gcn2) either globally or specifically in the liver display reduced capacity to maintain glucose homeostasis during fasting, suggesting the hypothesis that GCN2 may regulate gluconeogenesis (GNG), which normally plays a key role maintaining peripheral glucose homeostasis. Gcn2-deficient mice exhibit normal insulin sensitivity and plasma insulin but show reduced GNG when administered pyruvate, a gluconeogenic substrate. The basal expression of phosphoenolpyruvate carboxykinase, a rate-limiting enzyme in GNG, is abnormally elevated in Gcn2 knockout (KO) mice in the fed state but fails to be further induced during fasting. The level of tricarboxylic acid cycle intermediates, including malate and oxaloacetate, and the NADH-to-NAD(+) ratio are perturbed in the liver of Gcn2 KO mice either in the fed or fasted state, which may directly impinge upon GNG. Additionally, the expression of the CCAAT enhancer-binding protein-β (C/EBPβ) in the liver fails to be induced in Gcn2 KO mice after 24 h fasting, and the liver-specific Cebpβ KO mice show reduced fasting GNG similar to that seen in Gcn2-deficient mice. Our study demonstrates that GCN2 is important in maintaining GNG in the liver, which is likely to be mediated through regulation of C/EBPβ.
Collapse
Affiliation(s)
- Xu Xu
- Department of Biology, Center for Cellular Dynamics, Pennsylvania State University, University Park, Pennsylvania
| | | | | | | |
Collapse
|
187
|
Levant B, Ozias MK, Guilford BL, Wright DE. Streptozotocin-induced diabetes partially attenuates the effects of a high-fat diet on liver and brain fatty acid composition in mice. Lipids 2013; 48:939-48. [PMID: 23893338 DOI: 10.1007/s11745-013-3817-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 07/12/2013] [Indexed: 02/08/2023]
Abstract
The current study addresses the effects of a high-fat diet on liver and brain fatty acid compositions and the interaction of that diet with diabetes in a type 1 mouse model. Adult, male, normal and streptozotocin-induced diabetic C57BL/6 mice were fed standard (14 % kcal from fat) or high-fat (54 % kcal from fat, hydrogenated vegetable shortening and corn oil) diets for 8 weeks. Liver and whole brain total phospholipid fatty acid compositions were then determined by TLC/GC. In the liver of non-diabetic mice, the high-fat diet increased the percentages of 18:1n-9, 20:4n-6, and 22:5n-6 and decreased 18:2n-6 and 22:6n-3. Diabetes increased 16:0 in liver, and decreased 18:1n-7 and 20:4n-6. The effects of the high-fat diet on liver phospholipids in diabetic mice were similar to those in non-diabetic mice, or were of smaller magnitude. In the brain, the high-fat diet increased 18:0 and 20:4n-6 of non-diabetic, but not diabetic mice. Brain 22:5n-6 acid was increased by the high-fat diet in both non-diabetic and diabetic mice, but this increase was smaller in diabetic mice. Diabetes alone did not alter the percentage of any individual fatty acid in brain. This indicates that the effects of a high-fat diet on liver and brain phospholipid fatty acid compositions are partially attenuated by concomitant hyperglycemia with hypoinsulinemia.
Collapse
Affiliation(s)
- Beth Levant
- Departments of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Mail Stop 1018, 3901 Rainbow Blvd., Kansas City, KS 66160, USA.
| | | | | | | |
Collapse
|
188
|
Mao Y, Mohan R, Zhang S, Tang X. MicroRNAs as pharmacological targets in diabetes. Pharmacol Res 2013; 75:37-47. [PMID: 23810798 DOI: 10.1016/j.phrs.2013.06.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/10/2013] [Accepted: 06/12/2013] [Indexed: 12/14/2022]
Abstract
Diabetes is characterized by high levels of blood glucose due to either the loss of insulin-producing beta-cells in the pancreas, leading to a deficiency of insulin in type 1 diabetes, or due to increased insulin resistance, leading to reduced insulin sensitivity and productivity in type 2 diabetes. There is an increasing need for new options to treat diabetes, especially type 2 diabetes at its early stages due to an ineffective control of its development in patients. Recently, a novel class of small noncoding RNAs, termed microRNAs (miRNAs), is found to play a key role as important transcriptional and posttranscriptional inhibitors of gene expression in fine-tuning the target messenger RNAs (mRNAs). miRNAs are implicated in the pathogenesis of diabetes and have become an intriguing target for therapeutic intervention. This review focuses on the dysregulated miRNAs discovered in various diabetic models and addresses the potential for miRNAs to be therapeutic targets in the treatment of diabetes.
Collapse
Affiliation(s)
- Yiping Mao
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931, United States
| | | | | | | |
Collapse
|
189
|
Abstract
New insights into the actions of the hormone glucagon are provided by a recent study in rodents, which shows that glucagon can suppress hepatic glucose production by acting through the mediobasal hypothalamic region of the brain. This central regulatory mechanism is impaired in rats fed a high-fat diet, suggesting that hypothalamic glucagon resistance may be relevant to the hyperglycemia observed in obesity, diabetes or both (pages 766–772).
Collapse
Affiliation(s)
- Dale S Edgerton
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical School, Nashville, Tennessee, USA
| | | |
Collapse
|
190
|
McCall AL, Farhy LS. Treating type 1 diabetes: from strategies for insulin delivery to dual hormonal control. MINERVA ENDOCRINOL 2013; 38:145-63. [PMID: 23732369 PMCID: PMC4220674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Type 1 diabetes is a disorder where slow destruction of pancreatic β-cells occurs through autoimmune mechanisms. The result is a progressive and ultimately complete lack of endogenous insulin. Due to β-cell lack, secondary abnormalities in glucagon and likely in incretins occur. These multiple hormonal abnormalities cause metabolic instability and extreme glycemic variability, which is the primary phenotype. As the disease progresses patients often develop hypoglycemia unawareness and defects in their counterregulatory defenses. Intensive insulin therapy may thus lead to 3-fold excess of severe hypoglycemia and severely hinder the effective and safe control of hyperglycemia. The main goal of the therapy for type 1 diabetes has long been physiological mimicry of normal insulin secretion based on monitoring which requires considerable effort and understanding of the underlying physiology. Attainment of this goal is challenged by the nature of the disease and our current lack of means to fully repair the abnormal endocrine pancreas interactive functions. As a result, various insulin preparations have been developed to partially compensate for the inability to deliver timely exogenous insulin directly to the portal/intrapancreatic circulation. It remains an ongoing task to identify the ideal routes and regimens of their delivery and potentially that of other hormones to restore the deficient and disordered hormonal environment of the pancreas to achieve a near normal metabolic state. Several recent technological advances help addressing these goals, including the rapid progress in insulin pumps, continuous glucose sensors, and ultimately the artificial pancreas closed-loop technology and the recent start of dual-hormone therapies.
Collapse
Affiliation(s)
- A L McCall
- Department of Medicine and Center for Diabetes Technology, University of Virginia, Charlottesville, VA 22908, USA.
| | | |
Collapse
|
191
|
He S, Wang D, Lu Y, Chen Y, Jin X, Wang C, Zhao J, Ren Y, Wang L, Li H, Cheng J. Increasing glucagon secretion could antagonize the action of exogenous insulin for glycemic control in streptozocin-induced diabetic rhesus monkeys. Exp Biol Med (Maywood) 2013; 238:385-91. [PMID: 23760004 DOI: 10.1177/1535370213477974] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Although intraislet insulin signaling is known to play a critical role in regulating glucagon secretion, it is unknown whether abnormal glucagon secretion influences the hypoglycemic effect of exogenous insulin with intraislet insulin deletion. We performed a longitudinal study using 16 streptozocin (STZ)-induced diabetic rhesus monkeys to explore α-cell function under the absence β-cells and to assess whether increasing glucagon secretion antagonizes the action of exogenous insulin for glycemic control. We found that although the α-cells were impaired and the basal secretion levels of glucagon decreased rapidly after STZ (80–90 mg/kg) administration, as based on long-term observation post-STZ injection, glucagon secretion and the number of α-cells were increased. Glycemic control was increasingly difficult, the insulin resistance (HOMA-IR) index was significantly higher, and the triglycerides (TG) levels were gradually decreased. Moreover, a significant correlation between the levels of glucagon and HOMA-IR was found. Under the long-term absence of β-cells, the inhibitory effect on α-cell activity is profoundly attenuated, leading to an increase in glucagon secretion and the amount of α-cells and even α-cell dysfunction. Increased glucagon levels have a serious impact on the insulin sensitivity in vivo and result in an antagonization of the hypoglycemic effect of exogenous insulin.
Collapse
Affiliation(s)
- Sirong He
- Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University
| | - Dan Wang
- Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University
| | - Yanrong Lu
- Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University
| | - Younan Chen
- Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University
| | - Xi Jin
- Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University
| | - Chengsi Wang
- Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University
| | - Jingming Zhao
- Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University
| | - Yan Ren
- Department of Endocrine, West China Hospital, Sichuan University, No.1 Keyuan 4th Road, Gao Peng Avenue
| | - Li Wang
- National Center for Safety Evaluation of Traditional Chinese Medicine,Chengdu, Sichuan 610041, PR China
| | - Hongxia Li
- National Center for Safety Evaluation of Traditional Chinese Medicine,Chengdu, Sichuan 610041, PR China
| | - Jingqiu Cheng
- Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University
| |
Collapse
|
192
|
Xiao J, Guo R, Fung ML, Liong EC, Tipoe GL. Therapeutic approaches to non-alcoholic fatty liver disease: past achievements and future challenges. Hepatobiliary Pancreat Dis Int 2013; 12:125-35. [PMID: 23558065 DOI: 10.1016/s1499-3872(13)60021-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a leading cause of chronic liver injury and mortality in Western countries and China. However, as to date, there is no direct and effective therapy for this disease. The aim of this review is to analyze the key progress and challenges of main current therapeutic approaches in NAFLD. DATA SOURCE We carried out a PubMed search of English-language articles relevant to NAFLD therapy. RESULTS There are two major therapeutic strategies for NAFLD treatment: (1) lifestyle interventions (including weight reduction, dietary modification and physical exercise) and (2) pharmaceutical therapies. Lifestyle interventions, particularly chronic and moderate intensity exercise, are the most effective and recognized clinical therapies for NAFLD. For pharmaceutical therapies, although their effects and mechanisms have been extensively investigated in laboratory studies, they still need further tests and investigations in clinical human trials. CONCLUSION Future advancement of NAFLD therapy should focus on the mechanistic studies on cell based and animal models and human clinical trials of exercise, as well as the combination of lifestyle intervention and pharmaceutical therapy specifically targeting main signaling pathways related to lipid metabolism, oxidative stress and inflammation.
Collapse
Affiliation(s)
- Jia Xiao
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | | | | | | | | |
Collapse
|
193
|
Boyda HN, Procyshyn RM, Pang CCY, Barr AM. Peripheral adrenoceptors: the impetus behind glucose dysregulation and insulin resistance. J Neuroendocrinol 2013; 25:217-28. [PMID: 23140239 DOI: 10.1111/jne.12002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 10/10/2012] [Accepted: 11/04/2012] [Indexed: 12/20/2022]
Abstract
It is now accepted that several pharmacological drug treatments trigger clinical manifestations of glucose dysregulation, such as hyperglycaemia, glucose intolerance and insulin resistance, in part through poorly understood mechanisms. Persistent sympathoadrenal activation is linked to glucose dysregulation and insulin resistance, both of which significantly increase the risk of emergent endocrinological disorders, including metabolic syndrome and type 2 diabetes mellitus. Through the use of targeted mutagenesis and pharmacological methods, preclinical and clinical research has confirmed physiological glucoregulatory roles for several peripheral α- and β-adrenoceptor subtypes. Adrenoceptor isoforms in the pancreas (α(2A) and β(2) ), skeletal muscle (α(1A) and β(2) ), liver (α(1A & B) and β(2) ) and adipose tissue (α(1A) and β(1 & 3) ) are convincing aetiological targets that account for both immediate and long-lasting alterations in blood glucose homeostasis. Because significant overlap exists between the therapeutic applications of numerous classes of drugs and their associated adverse side-effects, a better understanding of peripheral adrenoceptor-mediated glucose metabolism is thus warranted. Therefore, at the same time as providing a brief review of glucose homeostasis in the periphery, the present review addresses both functional and pathophysiological roles of the mammalian α(1) , α(2) , and β-adrenoceptor isoforms in whole-body glucose turnover. We highlight evidence relating to the clinical use of common adrenergic drugs and their impacts on glucose metabolism.
Collapse
Affiliation(s)
- H N Boyda
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Vancouver, Canada.
| | | | | | | |
Collapse
|
194
|
Pang J, Rhodes DH, Pini M, Akasheh RT, Castellanos KJ, Cabay RJ, Cooper D, Perretti M, Fantuzzi G. Increased adiposity, dysregulated glucose metabolism and systemic inflammation in Galectin-3 KO mice. PLoS One 2013; 8:e57915. [PMID: 23451284 PMCID: PMC3579848 DOI: 10.1371/journal.pone.0057915] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/27/2013] [Indexed: 12/14/2022] Open
Abstract
Obesity and type 2 diabetes are associated with increased production of Galectin-3 (Gal-3), a protein that modulates inflammation and clearance of glucose adducts. We used Lean and Diet-induced Obese (DIO) WT and Gal-3 KO mice to investigate the role of Gal-3 in modulation of adiposity, glucose metabolism and inflammation. Deficiency of Gal-3 lead to age-dependent development of excess adiposity and systemic inflammation, as indicated by elevated production of acute-phase proteins, number of circulating pro-inflammatory Ly6Chigh monocytes and development of neutrophilia, microcytic anemia and thrombocytosis in 20-week-old Lean and DIO male Gal-3 KO mice. This was associated with impaired fasting glucose, heightened response to a glucose tolerance test and reduced adipose tissue expression of adiponectin, Gal-12, ATGL and PPARγ, in the presence of maintained insulin sensitivity and hepatic expression of gluconeogenic enzymes in 20-week-old Gal-3 KO mice compared to their diet-matched WT controls. Expression of PGC-1α and FGF-21 in the liver of Lean Gal-3 KO mice was comparable to that observed in DIO animals. Impaired fasting glucose and altered responsiveness to a glucose load preceded development of excess adiposity and systemic inflammation, as demonstrated in 12-week-old Gal-3 KO mice. Finally, a role for the microflora in mediating the fasting hyperglycemia, but not the excessive response to a glucose load, of 12-week-old Gal-3 KO mice was demonstrated by administration of antibiotics. In conclusion, Gal-3 is an important modulator of glucose metabolism, adiposity and inflammation.
Collapse
Affiliation(s)
- Jingbo Pang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Davina H. Rhodes
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Maria Pini
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Rand T. Akasheh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Karla J. Castellanos
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Robert J. Cabay
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Dianne Cooper
- The William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Mauro Perretti
- The William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Giamila Fantuzzi
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
195
|
Abstract
Despite improvements in the pharmacodynamics of injectable insulin and better insulin delivery systems, glucose control remains suboptimal in the majority of individuals with Type 1 diabetes. Profound defects in the physiological processes that normally maintain glucose homeostasis contribute to the difficulty in achieving glycaemic targets. Non-insulin-based adjunct treatments offer a potential means of complementing intensive insulin therapy in Type 1 diabetes through addressing some of the physiological disturbances that result from endogenous β-cell destruction, particularly through preservation of β-cell mass and prevention of apoptosis, and suppression of α-cell glucagon release in the postprandial state. The former approach applies most readily to newly diagnosed C-peptide-positive Type 1 diabetes, while the latter to established C-peptide-negative Type 1 diabetes. This review focuses primarily on the clinical trial data available on the use of non-insulin-based therapies in longer-duration Type 1 diabetes. We conclude that metformin may prove useful in macrovascular disease reduction, while pramlintide, glucagon-like peptide-1 agonists, dipeptidyl peptidase-4 inhibitors and leptin co-therapies may reduce HbA(1c) , glucose variability, postprandial glucose excursions and body weight. These early studies are encouraging and offer novel and potentially very effective approaches to the treatment of Type 1 diabetes, but the evidence is largely restricted to small, often uncontrolled trials. As such, these therapies cannot be currently recommended for routine clinical practice. There is a clear need to support large, multi-centre randomized controlled trials designed to establish whether adjunct insulin therapy has a place in the modern management of Type 1 diabetes.
Collapse
Affiliation(s)
- P George
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School
| | | |
Collapse
|
196
|
Amigo I, Traba J, González-Barroso MM, Rueda CB, Fernández M, Rial E, Sánchez A, Satrústegui J, Del Arco A. Glucagon regulation of oxidative phosphorylation requires an increase in matrix adenine nucleotide content through Ca2+ activation of the mitochondrial ATP-Mg/Pi carrier SCaMC-3. J Biol Chem 2013; 288:7791-7802. [PMID: 23344948 DOI: 10.1074/jbc.m112.409144] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
It has been known for a long time that mitochondria isolated from hepatocytes treated with glucagon or Ca(2+)-mobilizing agents such as phenylephrine show an increase in their adenine nucleotide (AdN) content, respiratory activity, and calcium retention capacity (CRC). Here, we have studied the role of SCaMC-3/slc25a23, the mitochondrial ATP-Mg/Pi carrier present in adult mouse liver, in the control of mitochondrial AdN levels and respiration in response to Ca(2+) signals as a candidate target of glucagon actions. With the use of SCaMC-3 knock-out (KO) mice, we have found that the carrier is responsible for the accumulation of AdNs in liver mitochondria in a strictly Ca(2+)-dependent way with an S0.5 for Ca(2+) activation of 3.3 ± 0.9 μm. Accumulation of matrix AdNs allows a SCaMC-3-dependent increase in CRC. In addition, SCaMC-3-dependent accumulation of AdNs is required to acquire a fully active state 3 respiration in AdN-depleted liver mitochondria, although further accumulation of AdNs is not followed by increases in respiration. Moreover, glucagon addition to isolated hepatocytes increases oligomycin-sensitive oxygen consumption and maximal respiratory rates in cells derived from wild type, but not SCaMC-3-KO mice and glucagon administration in vivo results in an increase in AdN content, state 3 respiration and CRC in liver mitochondria in wild type but not in SCaMC-3-KO mice. These results show that SCaMC-3 is required for the increase in oxidative phosphorylation observed in liver mitochondria in response to glucagon and Ca(2+)-mobilizing agents, possibly by allowing a Ca(2+)-dependent accumulation of mitochondrial AdNs and matrix Ca(2+), events permissive for other glucagon actions.
Collapse
Affiliation(s)
- Ignacio Amigo
- Departmento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid-CSIC; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER) 28049 Madrid
| | - Javier Traba
- Departmento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid-CSIC; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER) 28049 Madrid
| | - M Mar González-Barroso
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas (CIB), Consejo Superior de Investigaciones Científicas 28040 Madrid
| | - Carlos B Rueda
- Departmento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid-CSIC; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER) 28049 Madrid
| | - Margarita Fernández
- Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense de Madrid 28040 Madrid
| | - Eduardo Rial
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas (CIB), Consejo Superior de Investigaciones Científicas 28040 Madrid
| | - Aránzazu Sánchez
- Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense de Madrid 28040 Madrid
| | - Jorgina Satrústegui
- Departmento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid-CSIC; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER) 28049 Madrid.
| | - Araceli Del Arco
- Área de Bioquímica, Centro Regional de Investigaciones Biomédicas, Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla La Mancha (UCLM), Avda. Carlos III s/n, Toledo 45071, Spain.
| |
Collapse
|
197
|
Dose selection using a semi-mechanistic integrated glucose-insulin-glucagon model: designing phase 2 trials for a novel oral glucokinase activator. J Pharmacokinet Pharmacodyn 2012; 40:53-65. [PMID: 23263772 DOI: 10.1007/s10928-012-9286-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 12/07/2012] [Indexed: 10/27/2022]
Abstract
Selecting dosing regimens for phase 2 studies for a novel glucokinase activator LY2599506 is challenging due to the difficulty in modeling and assessing hypoglycemia risk. A semi-mechanistic integrated glucose-insulin-glucagon (GIG) model was developed in NONMEM based on pharmacokinetic, glucose, insulin, glucagon, and meal data obtained from a multiple ascending dose study in patients with Type 2 diabetes mellitus treated with LY2599506 for up to 26 days. The series of differential equations from the NONMEM model was translated into an R script to prospectively predict 24-h glucose profiles following LY2599506 treatment for 3 months for a variety of doses and dosing regimens. The reduction in hemoglobin A1c (HbA1c) at the end of the 3-month treatment was estimated using a transit compartment model based on the simulated fasting glucose values. Two randomized phase 2 studies, one with fixed dosing and the other employing conditional dose titration were conducted. The simulation suggested that (1) Comparable HbA1c lowering with lower hypoglycemia risk occurs with titration compared to fixed-dosing; and (2) A dose range of 50-400 mg BID provides either greater efficacy or lower hypoglycemia incidence or both than glyburide. The predictions were in reasonable agreement with the observed clinical data. The model predicted HbA1c reduction and hypoglycemia risk provided the basis for the decision to focus on the dose-titration trial and for the selection of doses for the demonstration of superiority of LY2599506 to glyburide. The integrated GIG model represented a valuable tool for the evaluation of hypoglycemia incidence.
Collapse
|
198
|
Schneck KB, Zhang X, Bauer R, Karlsson MO, Sinha VP. Assessment of glycemic response to an oral glucokinase activator in a proof of concept study: application of a semi-mechanistic, integrated glucose-insulin-glucagon model. J Pharmacokinet Pharmacodyn 2012; 40:67-80. [PMID: 23263773 DOI: 10.1007/s10928-012-9287-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 12/07/2012] [Indexed: 02/05/2023]
Abstract
A proof of concept study was conducted to investigate the safety and tolerability of a novel oral glucokinase activator, LY2599506, during multiple dose administration to healthy volunteers and subjects with Type 2 diabetes mellitus (T2DM). To analyze the study data, a previously established semi-mechanistic integrated glucose-insulin model was extended to include characterization of glucagon dynamics. The model captured endogenous glucose and insulin dynamics, including the amplifying effects of glucose on insulin production and of insulin on glucose elimination, as well as the inhibitory influence of glucose and insulin on hepatic glucose production. The hepatic glucose production in the model was increased by glucagon and glucagon production was inhibited by elevated glucose concentrations. The contribution of exogenous factors to glycemic response, such as ingestion of carbohydrates in meals, was also included in the model. The effect of LY2599506 on glucose homeostasis in subjects with T2DM was investigated by linking a one-compartment, pharmacokinetic model to the semi-mechanistic, integrated glucose-insulin-glucagon system. Drug effects were included on pancreatic insulin secretion and hepatic glucose production. The relationships between LY2599506, glucose, insulin, and glucagon concentrations were described quantitatively and consequently, the improved understanding of the drug-response system could be used to support further clinical study planning during drug development, such as dose selection.
Collapse
Affiliation(s)
- Karen B Schneck
- Global PK/PD/Pharmacometrics, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA.
| | | | | | | | | |
Collapse
|
199
|
Berger JH, Charron MJ, Silver DL. Major facilitator superfamily domain-containing protein 2a (MFSD2A) has roles in body growth, motor function, and lipid metabolism. PLoS One 2012; 7:e50629. [PMID: 23209793 PMCID: PMC3510178 DOI: 10.1371/journal.pone.0050629] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 10/22/2012] [Indexed: 12/24/2022] Open
Abstract
The metabolic adaptations to fasting in the liver are largely controlled by the nuclear hormone receptor peroxisome proliferator-activated receptor alpha (PPARα), where PPARα upregulates genes encoding the biochemical pathway for β-oxidation of fatty acids and ketogenesis. As part of an effort to identify and characterize nutritionally regulated genes that play physiological roles in the adaptation to fasting, we identified Major facilitator superfamily domain-containing protein 2a (Mfsd2a) as a fasting-induced gene regulated by both PPARα and glucagon signaling in the liver. MFSD2A is a cell-surface protein homologous to bacterial sodium-melibiose transporters. Hepatic expression and turnover of MFSD2A is acutely regulated by fasting/refeeding, but expression in the brain is constitutive. Relative to wildtype mice, gene-targeted Mfsd2a knockout mice are smaller, leaner, and have decreased serum, liver and brown adipose triglycerides. Mfsd2a knockout mice have normal liver lipid metabolism but increased whole body energy expenditure, likely due to increased β-oxidation in brown adipose tissue and significantly increased voluntary movement, but surprisingly exhibited a form of ataxia. Together, these results indicate that MFSD2A is a nutritionally regulated gene that plays myriad roles in body growth and development, motor function, and lipid metabolism. Moreover, these data suggest that the ligand(s) that are transported by MFSD2A play important roles in these physiological processes and await future identification.
Collapse
Affiliation(s)
- Justin H. Berger
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Maureen J. Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Obstetrics and Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - David L. Silver
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Signature Research Program in Cardiovascular & Metabolic Disorders, Duke-NUS Graduate Medical School, Singapore, Singapore
- * E-mail:
| |
Collapse
|
200
|
Langer S, Okar DA, Schultz J, Lenzen S, Baltrusch S. Dimer interface rearrangement of the 6-phosphofructo-2-kinase/fructose 2,6-bisphosphatase rat liver isoenzyme by cAMP-dependent Ser-32 phosphorylation. FEBS Lett 2012; 586:1419-25. [PMID: 22668829 DOI: 10.1016/j.febslet.2012.03.066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 03/07/2012] [Accepted: 03/31/2012] [Indexed: 10/28/2022]
Abstract
The bifunctional enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2/FBPase-2) is a key regulator of carbohydrate metabolism in liver. The goal of this study was to elucidate the regulatory role of Ser-32 phosphorylation on the kinase domain mediated dimerization of PFK-2/FBPase-2. Fluorescence-based mammalian two-hybrid and sensitized emission fluorescence resonance energy transfer analyses in cells revealed preferential binding within homodimers in contrast to heterodimers. Using isolated proteins a close proximity of two PFK-2/FBPase-2 monomers was only detectable in the phosphorylated enzyme dimer. Thus, a flexible kinase interaction mode exists, suggesting dimer conformation mediated coupling of hormonal and posttranslational enzyme regulation to the metabolic response in liver.
Collapse
Affiliation(s)
- Sara Langer
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | | | | | | | | |
Collapse
|