1
|
Accili D, Deng Z, Liu Q. Insulin resistance in type 2 diabetes mellitus. Nat Rev Endocrinol 2025:10.1038/s41574-025-01114-y. [PMID: 40247011 DOI: 10.1038/s41574-025-01114-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/28/2025] [Indexed: 04/19/2025]
Abstract
Insulin resistance is an integral pathophysiological feature of type 2 diabetes mellitus. Here, we review established and emerging cellular mechanisms of insulin resistance, their complex integrative features and their relevance to disease progression. While recognizing the heterogeneity of the elusive fundamental disruptions that cause insulin resistance, we endorse the view that effector mechanisms impinge on insulin receptor signalling and its relationship with plasma levels of insulin. We focus on hyperinsulinaemia and its consequences: acutely impaired but persistent insulin action, with reduced ability to lower glucose levels but preserved lipid synthesis and lipoprotein secretion. We emphasize the role of insulin sensitization as a therapeutic goal in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Domenico Accili
- Department of Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA.
| | - Zhaobing Deng
- Department of Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Qingli Liu
- Department of Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| |
Collapse
|
2
|
Al Harake SN, Abedin Y, Hatoum F, Nassar NZ, Ali A, Nassar A, Kanaan A, Bazzi S, Azar S, Harb F, Ghadieh HE. Involvement of a battery of investigated genes in lipid droplet pathophysiology and associated comorbidities. Adipocyte 2024; 13:2403380. [PMID: 39329369 PMCID: PMC11445895 DOI: 10.1080/21623945.2024.2403380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Lipid droplets (LDs) are highly specialized energy storage organelles involved in the maintenance of lipid homoeostasis by regulating lipid flux within white adipose tissue (WAT). The physiological function of adipocytes and LDs can be compromised by mutations in several genes, leading to NEFA-induced lipotoxicity, which ultimately manifests as metabolic complications, predominantly in the form of dyslipidemia, ectopic fat accumulation, and insulin resistance. In this review, we delineate the effects of mutations and deficiencies in genes - CIDEC, PPARG, BSCL2, AGPAT2, PLIN1, LIPE, LMNA, CAV1, CEACAM1, and INSR - involved in lipid droplet metabolism and their associated pathophysiological impairments, highlighting their roles in the development of lipodystrophies and metabolic dysfunction.
Collapse
Affiliation(s)
- Sami N. Al Harake
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Yasamin Abedin
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Fatema Hatoum
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Nour Zahraa Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Ali Ali
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Aline Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Amjad Kanaan
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Samer Bazzi
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Sami Azar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Frederic Harb
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Hilda E. Ghadieh
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| |
Collapse
|
3
|
Shrestha J, Limbu KR, Chhetri RB, Paudel KR, Hansbro PM, Oh YS, Baek DJ, Ki SH, Park EY. Antioxidant genes in cancer and metabolic diseases: Focusing on Nrf2, Sestrin, and heme oxygenase 1. Int J Biol Sci 2024; 20:4888-4907. [PMID: 39309448 PMCID: PMC11414382 DOI: 10.7150/ijbs.98846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
Reactive oxygen species are involved in the pathogenesis of cancers and metabolic diseases, including diabetes, obesity, and fatty liver disease. Thus, inhibiting the generation of free radicals is a promising strategy to control the onset of metabolic diseases and cancer progression. Various synthetic drugs and natural product-derived compounds that exhibit antioxidant activity have been reported to have a protective effect against a range of metabolic diseases and cancer. This review highlights the development and aggravation of cancer and metabolic diseases due to the imbalance between pro-oxidants and endogenous antioxidant molecules. In addition, we discuss the function of proteins that regulate the production of reactive oxygen species as a strategy to treat metabolic diseases. In particular, we summarize the role of proteins such as nuclear factor-like 2, Sestrin, and heme oxygenase-1, which regulate the expression of various antioxidant genes in metabolic diseases and cancer. We have included recent literature to discuss the latest research on identifying novel signals of antioxidant genes that can control metabolic diseases and cancer.
Collapse
Affiliation(s)
- Jitendra Shrestha
- College of Pharmacy, Mokpo National University, Jeonnam 58554, Republic of Korea
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Khem Raj Limbu
- College of Pharmacy, Mokpo National University, Jeonnam 58554, Republic of Korea
| | | | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sci., Sydney, NSW 2007, Australia
| | - Philip M. Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sci., Sydney, NSW 2007, Australia
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, Seongnam 13135, Republic of Korea
| | - Dong Jae Baek
- College of Pharmacy, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Sung-Hwan Ki
- College of Pharmacy, Chosun University, Gwangju 61451, Republic of Korea
| | - Eun-Young Park
- College of Pharmacy, Mokpo National University, Jeonnam 58554, Republic of Korea
| |
Collapse
|
4
|
Song J, Ni Q, Sun J, Xie J, Liu J, Ning G, Wang W, Wang Q. Aging Impairs Adaptive Unfolded Protein Response and Drives Beta Cell Dedifferentiation in Humans. J Clin Endocrinol Metab 2022; 107:3231-3241. [PMID: 36125175 PMCID: PMC9693768 DOI: 10.1210/clinem/dgac535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Diabetes is an age-related disease; however, the mechanism underlying senescent beta cell failure is still unknown. OBJECTIVE The present study was designed to investigate whether and how the differentiated state was altered in senescent human beta cells by excluding the effects of impaired glucose tolerance. METHODS We calculated the percentage of hormone-negative/chromogranin A-positive endocrine cells and evaluated the expressions of forkhead box O1 (FoxO1) and Urocortin 3 (UCN3) in islets from 31 nondiabetic individuals, divided into young (<40 years), middle-aged (40-60 years) and elderly (>60 years) groups. We also assessed adaptive unfolded protein response markers glucose-regulated protein 94 (GRP94), and spliced X-box binding protein 1 (XBP1s) in senescent beta cells and their possible contributions to maintaining beta cell identity and differentiation state. RESULTS We found an almost 2-fold increase in the proportion of dedifferentiated cells in elderly and middle-aged groups compared with the young group (3.1 ± 1.0% and 3.0 ± 0.9% vs 1.7 ± 0.5%, P < .001). This was accompanied by inactivation of FoxO1 and loss of UCN3 expression in senescent human beta cells. In addition, we demonstrated that the expression levels of adaptive unfolded protein response (UPR) components GRP94 and XBP1s declined with age. In vitro data showed knockdown GRP94 in Min6-triggered cells to dedifferentiate and acquire progenitor features, while restored GRP94 levels in H2O2-induced senescent Min6 cells rescued beta cell identity. CONCLUSION Our finding highlights that the failure to establish proper adaptive UPR in senescent human beta cells shifts their differentiated states, possibly representing a crucial step in the pathogenesis of age-related beta cell failure.
Collapse
Affiliation(s)
| | | | - Jiajun Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Xie
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianmin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Correspondence: Qidi Wang, MD, PhD, Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. ; or Weiqing Wang, MD, PhD, Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qidi Wang
- Correspondence: Qidi Wang, MD, PhD, Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. ; or Weiqing Wang, MD, PhD, Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
De Iuliis A, Montinaro E, Fatati G, Plebani M, Colosimo C. Diabetes mellitus and Parkinson's disease: dangerous liaisons between insulin and dopamine. Neural Regen Res 2022; 17:523-533. [PMID: 34380882 PMCID: PMC8504381 DOI: 10.4103/1673-5374.320965] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/08/2021] [Accepted: 03/04/2021] [Indexed: 11/13/2022] Open
Abstract
The relationship between diabetes mellitus and Parkinson's disease has been described in several epidemiological studies over the 1960s to date. Molecular studies have shown the possible functional link between insulin and dopamine, as there is strong evidence demonstrating the action of dopamine in pancreatic islets, as well as the insulin effects on feeding and cognition through central nervous system mechanism, largely independent of glucose utilization. Therapies used for the treatment of type 2 diabetes mellitus appear to be promising candidates for symptomatic and/or disease-modifying action in neurodegenerative diseases including Parkinson's disease, while an old dopamine agonist, bromocriptine, has been repositioned for the type 2 diabetes mellitus treatment. This review will aim at reappraising the different studies that have highlighted the dangerous liaisons between diabetes mellitus and Parkinson's disease.
Collapse
Affiliation(s)
| | - Ennio Montinaro
- Department of Neurology, Santa Maria University Hospital, Terni, Italy
| | | | - Mario Plebani
- Department of Medicine-DiMED, University of Padova, Italy
- Department of Medicine-DiMED, University of Padova, Padova, Italy; Department of Laboratory Medicine-Hospital of Padova, Padova, Italy
| | - Carlo Colosimo
- Department of Neurology, Santa Maria University Hospital, Terni, Italy
| |
Collapse
|
6
|
Hu XY, Chang Y, Xu ZZ, Wang Y, Dai MM, Yu KK, Sun CB, Dong MX, Zhang JX, Xu N, Liu WS, Chen ZA. Rubusoside Reduces Blood Glucose and Inhibits Oxidative Stress by Activating the AMPK Signaling Pathway in Type 2 Diabetes Mellitus Mice. Nat Prod Commun 2022. [DOI: 10.1177/1934578x211069230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The current study aimed at investigating the therapeutic effects of rubusoside on type 2 diabetes mellitus (T2DM) mice models as an alternative hypoglycemic candidate drug. T2DM mice models were established with a combination of streptozotocin (STZ) intraperitoneal injection and high-fat diet. After 10 weeks of rubusoside intragastric administration (100, 200 mg/kg/day) to the mice, the body weight, fasting blood glucose, glucose tolerance, and blood lipids were measured. The liver protein expression levels of p-AMPK, GLUT2, GLUT4 and total antioxidant capacity were also investigated. After 10 weeks of rubusoside administration, the levels of blood glucose and lipids were decreased in T2DM mice. Compared with the model group, rubusoside administration significantly decreased the liver mass-to-body weight ratio, upregulated p-AMPK and GLUT4, and downregulated GLUT2 expression levels in the liver. Activities of superoxide dismutase (SOD), catalase (CAT), and gluathione peroxidase (GSH-Px) were increased, and the concentration of malondialdehyde (MDA) was decreased to reduce oxidative stress in the liver. Liver hematoxylin and eosin (H&E) pathological analysis also showed that rubusoside had a protective effect on T2DM mice liver. These results demonstrate that rubusoside could be used as an anti-diabetic candidate drug, and that its hypoglycemic mechanism might be related to the activation of adenosine 5′-monophosphate (AMP)-activated protein kinase (AMPK) to modulate the expression of GLUT2 and GLUT4. Finally, rubusoside could also increase total antioxidant capacity to protect the liver from oxidative stress.
Collapse
Affiliation(s)
- Xi-yu Hu
- College of Medical, Yanbian University, Yanji, PR China
| | | | - Zheng-zhe Xu
- Affiliated Hospital of Yanbian University, Yanji Jilin 133002, China
| | - Yan Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, 130122, China
| | - Min-min Dai
- College of Medical, Yanbian University, Yanji, PR China
| | - Kai-kai Yu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, 130122, China
| | - Cheng-biao Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, 130122, China
| | - Ming-xin Dong
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, 130122, China
| | - Jian-xu Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, 130122, China
| | - Na Xu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, 130122, China
- Jilin Medical College, Jilin, China
| | - Wen-sen Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, 130122, China
| | - Zheng-ai Chen
- College of Medical, Yanbian University, Yanji, PR China
| |
Collapse
|
7
|
TOX4, an insulin receptor-independent regulator of hepatic glucose production, is activated in diabetic liver. Cell Metab 2022; 34:158-170.e5. [PMID: 34914893 PMCID: PMC8732315 DOI: 10.1016/j.cmet.2021.11.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/12/2021] [Accepted: 11/18/2021] [Indexed: 01/07/2023]
Abstract
Increased hepatic glucose production (HGP) contributes to hyperglycemia in type 2 diabetes. Hormonal regulation of this process is primarily, but not exclusively, mediated by the AKT-FoxO1 pathway. Here, we show that cAMP and dexamethasone regulate the high-mobility group superfamily member TOX4 to mediate HGP, independent of the insulin receptor/FoxO1 pathway. TOX4 inhibition decreases glucose production in primary hepatocytes and liver and increases glucose tolerance. Combined genetic ablation of TOX4 and FoxO1 in liver has additive effects on glucose tolerance and gluconeogenesis. Moreover, TOX4 ablation fails to reverse the metabolic derangement brought by insulin receptor knockout. TOX4 expression is increased in livers of patients with steatosis and diabetes and in diet-induced obese and db/db mice. In the latter two murine models, knockdown Tox4 decreases glycemia and improves glucose tolerance. We conclude that TOX4 is an insulin receptor-independent regulator of HGP and a candidate contributor to the pathophysiology of diabetes.
Collapse
|
8
|
Reilly AM, Yan S, Huang M, Abhyankar SD, Conley JM, Bone RN, Stull ND, Horan DJ, Roh HC, Robling AG, Ericsson AC, Dong XC, Evans-Molina C, Ren H. A high-fat diet catalyzes progression to hyperglycemia in mice with selective impairment of insulin action in Glut4-expressing tissues. J Biol Chem 2021; 298:101431. [PMID: 34801552 PMCID: PMC8689209 DOI: 10.1016/j.jbc.2021.101431] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/09/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022] Open
Abstract
Insulin resistance impairs postprandial glucose uptake through glucose transporter type 4 (GLUT4) and is the primary defect preceding type 2 diabetes. We previously generated an insulin-resistant mouse model with human GLUT4 promoter-driven insulin receptor knockout (GIRKO) in the muscle, adipose, and neuronal subpopulations. However, the rate of diabetes in GIRKO mice remained low prior to 6 months of age on normal chow diet (NCD), suggesting that additional factors/mechanisms are responsible for adverse metabolic effects driving the ultimate progression of overt diabetes. In this study, we characterized the metabolic phenotypes of the adult GIRKO mice acutely switched to high-fat diet (HFD) feeding in order to identify additional metabolic challenges required for disease progression. Distinct from other diet-induced obesity (DIO) and genetic models (e.g., db/db mice), GIRKO mice remained leaner on HFD feeding, but developed other cardinal features of insulin resistance syndrome. GIRKO mice rapidly developed hyperglycemia despite compensatory increases in β-cell mass and hyperinsulinemia. Furthermore, GIRKO mice also had impaired oral glucose tolerance and a limited glucose-lowering benefit from exendin-4, suggesting that the blunted incretin effect contributed to hyperglycemia. Secondly, GIRKO mice manifested severe dyslipidemia while on HFD due to elevated hepatic lipid secretion, serum triglyceride concentration, and lipid droplet accumulation in hepatocytes. Thirdly, GIRKO mice on HFD had increased inflammatory cues in the gut, which were associated with the HFD-induced microbiome alterations and increased serum lipopolysaccharide (LPS). In conclusion, our studies identified important gene/diet interactions contributing to diabetes progression, which might be leveraged to develop more efficacious therapies.
Collapse
Affiliation(s)
- Austin M Reilly
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Shijun Yan
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Menghao Huang
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Surabhi D Abhyankar
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jason M Conley
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert N Bone
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Natalie D Stull
- Indiana Biosciences Research Institute, Indianapolis, Indiana, USA
| | - Daniel J Horan
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hyun C Roh
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Alexander G Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Aaron C Ericsson
- Metagenomics Center, University of Missouri, Columbia, Missouri, USA
| | - Xiaocheng C Dong
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Carmella Evans-Molina
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA; Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
| | - Hongxia Ren
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, Indiana, USA; Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
9
|
Shankar K, Takemi S, Gupta D, Varshney S, Mani BK, Osborne-Lawrence S, Metzger NP, Richard CP, Berglund ED, Zigman JM. Ghrelin cell-expressed insulin receptors mediate meal- and obesity-induced declines in plasma ghrelin. JCI Insight 2021; 6:e146983. [PMID: 34473648 PMCID: PMC8492315 DOI: 10.1172/jci.insight.146983] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 08/04/2021] [Indexed: 01/20/2023] Open
Abstract
Mechanisms underlying postprandial and obesity-associated plasma ghrelin reductions are incompletely understood. Here, using ghrelin cell-selective insulin receptor-KO (GhIRKO) mice, we tested the impact of insulin, acting via ghrelin cell-expressed insulin receptors (IRs), to suppress ghrelin secretion. Insulin reduced ghrelin secretion from cultured gastric mucosal cells of control mice but not from those of GhIRKO mice. Acute insulin challenge and insulin infusion during both hyperinsulinemic-hypoglycemic clamps and hyperinsulinemic-euglycemic clamps lowered plasma ghrelin in control mice but not GhIRKO mice. Thus, ghrelin cell-expressed IRs are required for insulin-mediated reductions in plasma ghrelin. Furthermore, interventions that naturally raise insulin (glucose gavage, refeeding following fasting, and chronic high-fat diet) also lowered plasma ghrelin only in control mice - not GhIRKO mice. Thus, meal- and obesity-associated increases in insulin, acting via ghrelin cell-expressed IRs, represent a major, direct negative modulator of ghrelin secretion in vivo, as opposed to ingested or metabolized macronutrients. Refed GhIRKO mice exhibited reduced plasma insulin, highlighting ghrelin's actions to inhibit insulin release via a feedback loop. Moreover, GhIRKO mice required reduced glucose infusion rates during hyperinsulinemic-hypoglycemic clamps, suggesting that suppressed ghrelin release resulting from direct insulin action on ghrelin cells usually limits ghrelin's full potential to protect against insulin-induced hypoglycemia.
Collapse
Affiliation(s)
- Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Shota Takemi
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Sakuraku, Saitama, Japan
| | - Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Salil Varshney
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Bharath K. Mani
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Nathan P. Metzger
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Corine P. Richard
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Eric D. Berglund
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jeffrey M. Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
- Division of Endocrinology, Department of Internal Medicine, and
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
10
|
Rebelos E, Rinne JO, Nuutila P, Ekblad LL. Brain Glucose Metabolism in Health, Obesity, and Cognitive Decline-Does Insulin Have Anything to Do with It? A Narrative Review. J Clin Med 2021; 10:jcm10071532. [PMID: 33917464 PMCID: PMC8038699 DOI: 10.3390/jcm10071532] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 12/13/2022] Open
Abstract
Imaging brain glucose metabolism with fluorine-labelled fluorodeoxyglucose ([18F]-FDG) positron emission tomography (PET) has long been utilized to aid the diagnosis of memory disorders, in particular in differentiating Alzheimer’s disease (AD) from other neurological conditions causing cognitive decline. The interest for studying brain glucose metabolism in the context of metabolic disorders has arisen more recently. Obesity and type 2 diabetes—two diseases characterized by systemic insulin resistance—are associated with an increased risk for AD. Along with the well-defined patterns of fasting [18F]-FDG-PET changes that occur in AD, recent evidence has shown alterations in fasting and insulin-stimulated brain glucose metabolism also in obesity and systemic insulin resistance. Thus, it is important to clarify whether changes in brain glucose metabolism are just an epiphenomenon of the pathophysiology of the metabolic and neurologic disorders, or a crucial determinant of their pathophysiologic cascade. In this review, we discuss the current knowledge regarding alterations in brain glucose metabolism, studied with [18F]-FDG-PET from metabolic disorders to AD, with a special focus on how manipulation of insulin levels affects brain glucose metabolism in health and in systemic insulin resistance. A better understanding of alterations in brain glucose metabolism in health, obesity, and neurodegeneration, and the relationships between insulin resistance and central nervous system glucose metabolism may be an important step for the battle against metabolic and cognitive disorders.
Collapse
Affiliation(s)
- Eleni Rebelos
- Turku PET Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (E.R.); (J.O.R.); (P.N.)
| | - Juha O. Rinne
- Turku PET Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (E.R.); (J.O.R.); (P.N.)
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (E.R.); (J.O.R.); (P.N.)
- Department of Endocrinology, Turku University Hospital, 20520 Turku, Finland
| | - Laura L. Ekblad
- Turku PET Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland; (E.R.); (J.O.R.); (P.N.)
- Correspondence: ; Tel.: +358-2-3138721
| |
Collapse
|
11
|
Li G, Peng H, Qian S, Zou X, Du Y, Wang Z, Zou L, Feng Z, Zhang J, Zhu Y, Liang H, Li B. Bone Marrow-Derived Mesenchymal Stem Cells Restored High-Fat-Fed Induced Hyperinsulinemia in Rats at Early Stage of Type 2 Diabetes Mellitus. Cell Transplant 2021; 29:963689720904628. [PMID: 32228047 PMCID: PMC7502689 DOI: 10.1177/0963689720904628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Numerous studies have proposed the transplantation of mesenchymal stem cells
(MSCs) in the treatment of typical type 2 diabetes mellitus (T2DM). We aimed to
find a new strategy with MSC therapy at an early stage of T2DM to efficiently
prevent the progressive deterioration of organic dysfunction. Using the
high-fat-fed hyperinsulinemia rat model, we found that before the onset of
typical T2DM, bone marrow-derived MSCs (BM-MSCs) significantly attenuated rising
insulin with decline in glucose as well as restored lipometabolic disorder and
liver dysfunction. BM-MSCs also favored the histological structure recovery and
proliferative capacity of pancreatic islet cells. More importantly, BM-MSC
administration successfully reversed the abnormal expression of insulin
resistance-related proteins including GLUT4, phosphorylated insulin receptor
substrate 1, and protein kinase Akt and proinflammatory cytokines IL-6 and TNFα
in liver. These findings suggested that MSCs transplantation during
hyperinsulinemia could prevent most potential risks of T2DM for patients.
Collapse
Affiliation(s)
- Gongchi Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Peng
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Shen Qian
- School of Foreign Studies of Zhongnan University of Economics and Law, Wuhan, China
| | - Xinhua Zou
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Chronic Wound and Diabetic Foot Clinical Research Center, Wuhan, China
| | - Ye Du
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Chronic Wound and Diabetic Foot Clinical Research Center, Wuhan, China
| | - Zhi Wang
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Chronic Wound and Diabetic Foot Clinical Research Center, Wuhan, China
| | - Lijun Zou
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Chronic Wound and Diabetic Foot Clinical Research Center, Wuhan, China
| | - Zibo Feng
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Chronic Wound and Diabetic Foot Clinical Research Center, Wuhan, China
| | - Jing Zhang
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Chronic Wound and Diabetic Foot Clinical Research Center, Wuhan, China
| | - Youpeng Zhu
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Chronic Wound and Diabetic Foot Clinical Research Center, Wuhan, China
| | - Huamin Liang
- Department of Physiology, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China.,Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China
| | - Binghui Li
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Chronic Wound and Diabetic Foot Clinical Research Center, Wuhan, China
| |
Collapse
|
12
|
Huang Q, Huang Y, Liu J. Mesenchymal Stem Cells: An Excellent Candidate for the Treatment of Diabetes Mellitus. Int J Endocrinol 2021; 2021:9938658. [PMID: 34135959 PMCID: PMC8178013 DOI: 10.1155/2021/9938658] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells (ASCs) known for repairing damaged cells, exerting anti-inflammatory responses and producing immunoregulatory effects that can be significantly induced into insulin-producing cells (IPCs), providing an inexhaustible supply of functional β cells for cell replacement therapy and disease modeling for diabetes. MSC therapy may be the most promising strategy for diabetes mellitus because of these significant merits. In this paper, we focused on MSC therapy for diabetes.
Collapse
Affiliation(s)
- Qiulan Huang
- Department of Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yanting Huang
- Department of Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jianping Liu
- Department of Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
13
|
Xiong J, Hu H, Guo R, Wang H, Jiang H. Mesenchymal Stem Cell Exosomes as a New Strategy for the Treatment of Diabetes Complications. Front Endocrinol (Lausanne) 2021; 12:646233. [PMID: 33995278 PMCID: PMC8117220 DOI: 10.3389/fendo.2021.646233] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/12/2021] [Indexed: 01/01/2023] Open
Abstract
Diabetes mellitus (DM) is a metabolic disease, now prevalent worldwide, which is characterized by a relative or absolute lack of insulin secretion leading to chronically increased blood glucose levels. Diabetic patients are often accompanied by multiple macrovascular complications, such as coronary heart disease, hypertension, macrovascular arteriosclerosis, and microvascular complications. Microvascular complications include diabetic kidney injury, diabetic encephalopathy, and diabetic foot, which reduce the quality of life and survival status of patients. Mesenchymal stem cell exosomes (MSC-Exos) possess repair functions similar to MSCs, low immunogenicity, and ease of storage and transport. MSC-Exos have been proven to possess excellent repair effects in repairing various organ damages. This study reviews the application of MSC-Exos in the treatment of DM and its common complications. MSC-Exos may be used as an effective treatment for DM and its complications.
Collapse
Affiliation(s)
| | | | | | - Hui Wang
- *Correspondence: Hui Wang, ; Hua Jiang,
| | - Hua Jiang
- *Correspondence: Hui Wang, ; Hua Jiang,
| |
Collapse
|
14
|
Fan J, Du W, Kim-Muller JY, Son J, Kuo T, Larrea D, Garcia C, Kitamoto T, Kraakman MJ, Owusu-Ansah E, Cirulli V, Accili D. Cyb5r3 links FoxO1-dependent mitochondrial dysfunction with β-cell failure. Mol Metab 2020; 34:97-111. [PMID: 32180563 PMCID: PMC7031142 DOI: 10.1016/j.molmet.2019.12.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/03/2019] [Accepted: 12/12/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Diabetes is characterized by pancreatic β-cell dedifferentiation. Dedifferentiating β cells inappropriately metabolize lipids over carbohydrates and exhibit impaired mitochondrial oxidative phosphorylation. However, the mechanism linking the β-cell's response to an adverse metabolic environment with impaired mitochondrial function remains unclear. METHODS Here we report that the oxidoreductase cytochrome b5 reductase 3 (Cyb5r3) links FoxO1 signaling to β-cell stimulus/secretion coupling by regulating mitochondrial function, reactive oxygen species generation, and nicotinamide actin dysfunction (NAD)/reduced nicotinamide actin dysfunction (NADH) ratios. RESULTS The expression of Cyb5r3 is decreased in FoxO1-deficient β cells. Mice with β-cell-specific deletion of Cyb5r3 have impaired insulin secretion, resulting in glucose intolerance and diet-induced hyperglycemia. Cyb5r3-deficient β cells have a blunted respiratory response to glucose and display extensive mitochondrial and secretory granule abnormalities, consistent with altered differentiation. Moreover, FoxO1 is unable to maintain expression of key differentiation markers in Cyb5r3-deficient β cells, suggesting that Cyb5r3 is required for FoxO1-dependent lineage stability. CONCLUSIONS The findings highlight a pathway linking FoxO1 to mitochondrial dysfunction that can mediate β-cell failure.
Collapse
Affiliation(s)
- Jason Fan
- Naomi Berrie Diabetes Center and Departments of Medicine, Columbia University, New York, NY 10032, USA
| | - Wen Du
- Naomi Berrie Diabetes Center and Departments of Medicine, Columbia University, New York, NY 10032, USA
| | - Ja Young Kim-Muller
- Naomi Berrie Diabetes Center and Departments of Medicine, Columbia University, New York, NY 10032, USA
| | - Jinsook Son
- Naomi Berrie Diabetes Center and Departments of Medicine, Columbia University, New York, NY 10032, USA
| | - Taiyi Kuo
- Naomi Berrie Diabetes Center and Departments of Medicine, Columbia University, New York, NY 10032, USA
| | - Delfina Larrea
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Christian Garcia
- Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
| | - Takumi Kitamoto
- Naomi Berrie Diabetes Center and Departments of Medicine, Columbia University, New York, NY 10032, USA
| | - Michael J Kraakman
- Naomi Berrie Diabetes Center and Departments of Medicine, Columbia University, New York, NY 10032, USA
| | - Edward Owusu-Ansah
- Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
| | - Vincenzo Cirulli
- Department of Medicine, UW-Diabetes Institute, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Domenico Accili
- Naomi Berrie Diabetes Center and Departments of Medicine, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
15
|
Nasoohi S, Parveen K, Ishrat T. Metabolic Syndrome, Brain Insulin Resistance, and Alzheimer's Disease: Thioredoxin Interacting Protein (TXNIP) and Inflammasome as Core Amplifiers. J Alzheimers Dis 2019; 66:857-885. [PMID: 30372683 DOI: 10.3233/jad-180735] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Empirical evidence indicates a strong association between insulin resistance and pathological alterations related to Alzheimer's disease (AD) in different cerebral regions. While cerebral insulin resistance is not essentially parallel with systemic metabolic derangements, type 2 diabetes mellitus (T2DM) has been established as a risk factor for AD. The circulating "toxic metabolites" emerging in metabolic syndrome may engage several biochemical pathways to promote oxidative stress and neuroinflammation leading to impair insulin function in the brain or "type 3 diabetes". Thioredoxin-interacting protein (TXNIP) as an intracellular amplifier of oxidative stress and inflammasome activation may presumably mediate central insulin resistance. Emerging data including those from our recent studies has demonstrated a sharp TXNIP upregulation in stroke, aging and AD and well underlining the significance of this hypothesis. With the main interest to illustrate TXNIP place in type 3 diabetes, the present review primarily briefs the potential mechanisms contributing to cerebral insulin resistance in a metabolically deranged environment. Then with a particular focus on plausible TXNIP functions to drive and associate with AD pathology, we present the most recent evidence supporting TXNIP as a promising therapeutic target in AD as an age-associated dementia.
Collapse
|
16
|
Ren H, Vieira-de-Abreu A, Yan S, Reilly AM, Chan O, Accili D. Altered Central Nutrient Sensing in Male Mice Lacking Insulin Receptors in Glut4-Expressing Neurons. Endocrinology 2019; 160:2038-2048. [PMID: 31199472 PMCID: PMC6691683 DOI: 10.1210/en.2019-00341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/08/2019] [Indexed: 12/20/2022]
Abstract
Insulin signaling in the central nervous system influences satiety, counterregulation, and peripheral insulin sensitivity. Neurons expressing the Glut4 glucose transporter influence peripheral insulin sensitivity. Here, we analyzed the effects of insulin receptor (IR) signaling in hypothalamic Glut4 neurons on glucose sensing as well as leptin and amino acid signaling. By measuring electrophysiological responses to low glucose conditions, we found that the majority of Glut4 neurons in the ventromedial hypothalamus (VMH) were glucose excitatory neurons. GLUT4-Cre-driven insulin receptor knockout mice with a combined ablation of IR in Glut4-expressing tissues showed increased counterregulatory response to either 2-deoxyglucose-induced neuroglycopenia or systemic insulin-induced hypoglycemia. The latter response was recapitulated in mice with decreased VMH IR expression, suggesting that the effects on the counterregulatory response are likely mediated through the deletion of IRs on Glut4 neurons in the VMH. Using immunohistochemistry in fluorescently labeled hypothalamic Glut4 neurons, we showed that IR signaling promoted hypothalamic cellular signaling responses to the rise of insulin, leptin, and amino acids associated with feeding. We concluded that hypothalamic Glut4 neurons modulated the glucagon counterregulatory response and that IR signaling in Glut4 neurons was required to integrate hormonal and nutritional cues for the regulation of glucose metabolism.
Collapse
Affiliation(s)
- Hongxia Ren
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
- Correspondence: Hongxia Ren, PhD, Indiana University School of Medicine, 635 Barnhill Drive, MS2031, Indianapolis, Indiana 46202. E-mail: ; or Owen Chan, PhD, Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah, 15 North 2030 East, EIHG Building 533, Room 2420B, Salt Lake City, Utah 84112. E-mail:
| | - Adriana Vieira-de-Abreu
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah, Salt Lake City, Utah
| | - Shijun Yan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Austin M Reilly
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Owen Chan
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah, Salt Lake City, Utah
- Correspondence: Hongxia Ren, PhD, Indiana University School of Medicine, 635 Barnhill Drive, MS2031, Indianapolis, Indiana 46202. E-mail: ; or Owen Chan, PhD, Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah, 15 North 2030 East, EIHG Building 533, Room 2420B, Salt Lake City, Utah 84112. E-mail:
| | - Domenico Accili
- Department of Medicine and Berrie Diabetes Center, Columbia University College of Physicians & Surgeons, New York, New York
| |
Collapse
|
17
|
Yang Y, Zhao M, Yu XJ, Liu LZ, He X, Deng J, Zang WJ. Pyridostigmine regulates glucose metabolism and mitochondrial homeostasis to reduce myocardial vulnerability to injury in diabetic mice. Am J Physiol Endocrinol Metab 2019; 317:E312-E326. [PMID: 31211620 DOI: 10.1152/ajpendo.00569.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diabetic patients are more susceptible to myocardial ischemia damage than nondiabetic patients, with worse clinical outcomes and greater mortality. The mechanism may be related to glucose metabolism, mitochondrial homeostasis, and oxidative stress. Pyridostigmine may improve vagal activity to protect cardiac function in cardiovascular diseases. Researchers have not determined whether pyridostigmine regulates glucose metabolism and mitochondrial homeostasis to reduce myocardial vulnerability to injury in diabetic mice. In the present study, autonomic imbalance, myocardial damage, mitochondrial dysfunction, and oxidative stress were exacerbated in isoproterenol-stimulated diabetic mice, revealing the myocardial vulnerability of diabetic mice to injury compared with mice with diabetes or exposed to isoproterenol alone. Compared with normal mice, the expression of glucose transporters (GLUT)1/4 phosphofructokinase (PFK) FB3, and pyruvate kinase isoform (PKM) was decreased in diabetic mice, but increased in isoproterenol-stimulated normal mice. Following exposure to isoproterenol, the expression of (GLUT)1/4 phosphofructokinase (PFK) FB3, and PKM decreased in diabetic mice compared with normal mice. The downregulation of SIRT3/AMPK and IRS-1/Akt in isoproterenol-stimulated diabetic mice was exacerbated compared with that in diabetic mice or isoproterenol-stimulated normal mice. Pyridostigmine improved vagus activity, increased GLUT1/4, PFKFB3, and PKM expression, and ameliorated mitochondrial dysfunction and oxidative stress to reduce myocardial damage in isoproterenol-stimulated diabetic mice. Based on these results, it was found that pyridostigmine may reduce myocardial vulnerability to injury via the SIRT3/AMPK and IRS-1/Akt pathways in diabetic mice with isoproterenol-induced myocardial damage. This study may provide a potential therapeutic target for myocardial damage in diabetic patients.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Ming Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Xiao-Jiang Yu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Long-Zhu Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Xi He
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Juan Deng
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Wei-Jin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| |
Collapse
|
18
|
de Groot MHM, Castorena CM, Cox KH, Kumar V, Mohawk JA, Ahmed NI, Takahashi JS. A novel mutation in Slc2a4 as a mouse model of fatigue. GENES BRAIN AND BEHAVIOR 2019; 18:e12578. [PMID: 31059591 DOI: 10.1111/gbb.12578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 11/28/2022]
Abstract
Chronic fatigue is a debilitating disorder with widespread consequences, but effective treatment strategies are lacking. Novel genetic mouse models of fatigue may prove invaluable for studying its underlying physiological mechanisms and for testing treatments and interventions. In a screen of voluntary wheel-running behavior in N-ethyl-N-nitrosourea mutagenized C57BL/6J mice, we discovered two lines with low body weights and aberrant wheel-running patterns suggestive of a fatigue phenotype. Affected progeny from these lines had lower daily activity levels and exhibited low amplitude circadian rhythm alterations. Their aberrant behavior was characterized by frequent interruptions and periods of inactivity throughout the dark phase of the light-dark cycle and increased levels of activity during the rest or light phase. Expression of the behavioral phenotypes in offspring of strategic crosses was consistent with a recessive inheritance pattern. Mapping of phenotypic abnormalities showed linkage with a single locus on chromosome 1, and whole exome sequencing identified a single point mutation in the Slc2a4 gene encoding the GLUT4 insulin-responsive glucose transporter. The single nucleotide change (A-T, which we named "twiggy") was in the distal end of exon 10 and resulted in a premature stop (Y440*). Additional metabolic phenotyping confirmed that these mice recapitulate phenotypes found in GLUT4 knockout mice. However, to the best of our knowledge, this is the first time a mutation in this gene has been shown to result in extensive changes in general behavioral patterns. These findings suggest that GLUT4 may be involved in circadian behavioral abnormalities and could provide insights into fatigue in humans.
Collapse
Affiliation(s)
- Marleen H M de Groot
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Carlos M Castorena
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kimberly H Cox
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Vivek Kumar
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jennifer A Mohawk
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Newaz I Ahmed
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Joseph S Takahashi
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
19
|
Insulin and Insulin Receptors in Adipose Tissue Development. Int J Mol Sci 2019; 20:ijms20030759. [PMID: 30754657 PMCID: PMC6387287 DOI: 10.3390/ijms20030759] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022] Open
Abstract
Insulin is a major endocrine hormone also involved in the regulation of energy and lipid metabolism via the activation of an intracellular signaling cascade involving the insulin receptor (INSR), insulin receptor substrate (IRS) proteins, phosphoinositol 3-kinase (PI3K) and protein kinase B (AKT). Specifically, insulin regulates several aspects of the development and function of adipose tissue and stimulates the differentiation program of adipose cells. Insulin can activate its responses in adipose tissue through two INSR splicing variants: INSR-A, which is predominantly expressed in mesenchymal and less-differentiated cells and mainly linked to cell proliferation, and INSR-B, which is more expressed in terminally differentiated cells and coupled to metabolic effects. Recent findings have revealed that different distributions of INSR and an altered INSR-A:INSR-B ratio may contribute to metabolic abnormalities during the onset of insulin resistance and the progression to type 2 diabetes. In this review, we discuss the role of insulin and the INSR in the development and endocrine activity of adipose tissue and the pharmacological implications for the management of obesity and type 2 diabetes.
Collapse
|
20
|
Sun J, Ni Q, Xie J, Xu M, Zhang J, Kuang J, Wang Y, Ning G, Wang Q. β-Cell Dedifferentiation in Patients With T2D With Adequate Glucose Control and Nondiabetic Chronic Pancreatitis. J Clin Endocrinol Metab 2019; 104:83-94. [PMID: 30085195 DOI: 10.1210/jc.2018-00968] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/31/2018] [Indexed: 12/15/2022]
Abstract
CONTEXT Type 2 diabetes (T2D) and pancreatogenic diabetes are both associated with loss of functional β-cell mass. Previous studies have proposed β-cell dedifferentiation as a mechanism of islet β-cell failure, but its significance in humans is still controversial. OBJECTIVE To determine whether β-cell dedifferentiation occurs in human T2D with adequate glucose control and in nondiabetic chronic pancreatitis (NDCP), we examined pancreatic islets from nine nondiabetic controls, 10 patients with diabetes with well-controlled fasting glycemia, and four individuals with NDCP. DESIGN We calculated the percentage of hormone-negative endocrine cells and multihormone endocrine cells and scored the pathological characteristics; that is, inflammatory cell infiltration, fibrosis, atrophy, and steatosis, in each case. RESULTS We found a nearly threefold increase in dedifferentiated cells in T2D with adequate glucose control compared with nondiabetic controls (10.0% vs 3.6%, T2D vs nondiabetic controls, P < 0.0001). The dedifferentiation rate was positively correlated with the duration of diabetes. Moreover, we detected a considerable proportion of dedifferentiated cells in NDCP (10.4%), which correlated well with the grade of inflammatory cell infiltration, fibrosis, and atrophy. CONCLUSIONS The data support the view that pancreatic β-cells are dedifferentiated in patients with T2D with adequate glucose control. Furthermore, the existence of abundant dedifferentiated cells in NDCP suggests that inflammation-induced β-cell dedifferentiation can be a cause of pancreatogenic diabetes during disease progress.
Collapse
Affiliation(s)
- Jiajun Sun
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qicheng Ni
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Xie
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Kuang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanqiu Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qidi Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Accili D. Insulin Action Research and the Future of Diabetes Treatment: The 2017 Banting Medal for Scientific Achievement Lecture. Diabetes 2018; 67:1701-1709. [PMID: 30135131 PMCID: PMC6110318 DOI: 10.2337/dbi18-0025] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetes is caused by combined abnormalities in insulin production and action. The pathophysiology of these defects has been studied extensively and is reasonably well understood. Their causes are elusive and their manifestations pleiotropic, likely reflecting the triple threat of genes, environment, and lifestyle. Treatment, once restricted to monotherapy with secretagogues or insulin, now involves complex combinations of expensive regimens that stem the progression but do not fundamentally alter the underlying causes of the disease. As advances in our understanding of insulin action and β-cell failure reach a critical stage, here I draw on lessons learned from our research on insulin regulation of gene expression and pancreatic β-cell dedifferentiation to address the question of how we can translate this exciting biology into mechanism-based interventions to reverse the course of diabetes.
Collapse
MESH Headings
- Animals
- Awards and Prizes
- Cell Dedifferentiation/drug effects
- Cell Transdifferentiation/drug effects
- Cellular Reprogramming/drug effects
- Combined Modality Therapy/adverse effects
- Diabetes Complications/prevention & control
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/therapy
- Drug Design
- Drug Therapy, Combination/adverse effects
- Enteroendocrine Cells/drug effects
- Enteroendocrine Cells/metabolism
- Enteroendocrine Cells/pathology
- Forkhead Transcription Factors/antagonists & inhibitors
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Humans
- Hypoglycemic Agents/adverse effects
- Hypoglycemic Agents/chemistry
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Insulin/adverse effects
- Insulin/metabolism
- Insulin/pharmacology
- Insulin/therapeutic use
- Insulin Resistance
- Insulin Secretion
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Models, Biological
Collapse
Affiliation(s)
- Domenico Accili
- Naomi Berrie Diabetes Center and Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
22
|
Hogg DW, Chen Y, D'Aquila AL, Xu M, Husić M, Tan LA, Bull C, Lovejoy DA. A novel role of the corticotrophin-releasing hormone regulating peptide, teneurin C-terminal associated peptide 1, on glucose uptake into the brain. J Neuroendocrinol 2018; 30:e12579. [PMID: 29411913 DOI: 10.1111/jne.12579] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 01/11/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023]
Abstract
Teneurin C-terminal associated peptide (TCAP) is an ancient paracrine signalling agent that evolved via lateral gene transfer from prokaryotes into an early metazoan ancestor. Although it bears structural similarity to corticotrophin-releasing hormone (CRH), it inhibits the in vivo actions of CRH. The TCAPs are highly expressed in neurones, where they induce rapid cytoskeletal rearrangement and are neuroprotective. Because these processes are highly energy-dependent, this suggests that TCAP has the potential to regulate glucose uptake because glucose is the primary energy substrate in brain, and neurones require a steady supply to meet the high metabolic demands of neuronal communication. Therefore, the objective of the present study was to assess the effect of TCAP-mediated glucose uptake in the brain and in neuronal cell models. TCAP-mediated 18 F-deoxyglucose (FDG) uptake into brain tissue was assessed in male wild-type Wistar rats by functional positron emission tomography. TCAP-1 increased FDG uptake by over 40% into cortical regions of the brain, demonstrating that TCAP-1 can significantly enhance glucose supply. Importantly, a single nanomolar injection of TCAP-1 increased brain glucose after 3 days and decreased blood glucose after 1 week. This is corroborated by a decreased serum concentration of insulin and an increased serum concentration of glucagon. In immortalised hypothalamic neurones, TCAP-1 increased ATP production and enhanced glucose uptake by increasing glucose transporter recruitment to the plasma membrane likely via AKT and mitogen-activated protein kinase/ERK phosphorylation events. Taken together, these data demonstrate that TCAP-1 increases glucose metabolism in neurones, and may represent a peptide signalling agent that regulated glucose uptake before insulin and related peptides.
Collapse
Affiliation(s)
- D W Hogg
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Y Chen
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - A L D'Aquila
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - M Xu
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - M Husić
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - L A Tan
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - C Bull
- Molecular Imaging Inc., Ann Arbor, MI, USA
| | - D A Lovejoy
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
23
|
Fakhoury H, Osman S, Ghazale N, Dahdah N, El-Sibai M, Kanaan A. Enhanced Glucose Uptake in Phenylbutyric Acid-Treated 3T3-L1 Adipocytes. CELL AND TISSUE BIOLOGY 2018; 12:48-56. [DOI: 10.1134/s1990519x18010066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Indexed: 01/02/2025]
|
24
|
Lee TI, Bai KJ, Chen YC, Lee TW, Chung CC, Tsai WC, Tsao SY, Kao YH. Histone deacetylase inhibition of cardiac autophagy in rats on a high‑fat diet with low‑dose streptozotocin-induced type 2 diabetes mellitus. Mol Med Rep 2017; 17:594-601. [PMID: 29115461 DOI: 10.3892/mmr.2017.7905] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 08/03/2017] [Indexed: 11/06/2022] Open
Abstract
Autophagy serves a role in preserving cellular homeostasis. Diabetes mellitus (DM) impairs cardiac autophagy and is associated with an accumulation of cytotoxic proteins that may provoke apoptosis and damage cardiomyocytes. Histone deacetylase (HDAC) inhibitors attenuate cardiac fibrosis and inflammation, and improve cardiomyopathy resulting from DM. However, the effect of HDAC inhibition on autophagy in DM cardiomyopathy has not been investigated. The purpose of the present study was to evaluate whether HDAC inhibition modulates cardiac autophagy and to investigate the potential mechanisms in type 2 DM (T2DM) hearts. Electrocardiography was used to evaluate cardiac function and western blotting was used to evaluate protein expression in autophagy, the serine/threonine protein kinase mTOR (mTOR) signaling pathway, poly adenosine diphosphate ribose polymerase 1 (PARP1), insulin signaling, advanced glycosylation end product‑specific receptor (RAGE), and proinflammatory cytokines in control rats and in rats treated with a high‑fat diet (60% fat) and low‑dose streptozotocin (35 mg/kg) in order to induce T2DM, with or without an HDAC inhibitor (MPT0E014; 50 mg/kg/rat daily for 7 days). Compared with the control rats, T2DM and T2DM rats treated with MPT0E014 exhibited elevated blood glucose levels and similar body weights. However, T2DM rats treated with MPT0E014 and control rats had a smaller left ventricular end‑diastolic diameter compared with the T2DM rats. The control and T2DM rats treated with MPT0E014 had greater protein expression of cardiac phosphorylated (p)‑5' adenosine monophosphate‑activated protein kinase α 2, light chain 3‑II, Beclin‑1, glucose transporter 4, p‑protein kinase B, and insulin receptor substrate‑1 (Ser 307) compared with T2DM rats. In addition, control and T2DM rats treated with MPT0E014 had decreased cardiac protein expression of cleaved PARP1, p‑mTOR‑S2448, p‑P70S6K‑Thr‑389, RAGE, tumor necrosis factor‑α, and interleukin‑6 compared with T2DM rats. The present study demonstrated that MPT0E014 may improve cardiac function in T2DM rats by modulating myocardial autophagy, inflammation and insulin signaling.
Collapse
Affiliation(s)
- Ting-I Lee
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| | - Kuan-Jen Bai
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan, R.O.C
| | - Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei 11696, Taiwan, R.O.C
| | - Cheng-Chih Chung
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| | - Wen-Chih Tsai
- Division of Cardiology, Tzu‑Chi General Hospital, Institute of Medical Sciences, Tzu‑Chi University, Hualien 97004, Taiwan, R.O.C
| | - Shin-Yi Tsao
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City 22174, Taiwan, R.O.C
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| |
Collapse
|
25
|
Wang W, Liu C, Jimenez-Gonzalez M, Song WJ, Hussain MA. The undoing and redoing of the diabetic β-cell. J Diabetes Complications 2017; 31:912-917. [PMID: 28242267 DOI: 10.1016/j.jdiacomp.2017.01.028] [cited] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/23/2016] [Accepted: 01/31/2017] [Indexed: 01/26/2025]
Abstract
A hallmark of type 2 diabetes (T2DM) is the reduction in functional β-cell mass, which is considered at least in part to result from an imbalance of β-cell renewal and apoptosis, with the latter being accelerated during metabolic stress. More recent studies, however, suggest that the loss of functional β-cell mass is not as much due to β-cell death but rather to de-differentiation of β-cells when these cells are exposed to metabolic stressors, opening the possibility to re-differentiate and restore functional β-cell mass by therapeutic intervention. In parallel, clinical observations suggest that temporary intensive insulin therapy in early diagnosed humans with T2DM, so as to "rest" endogenous β-cells, allows these patients to regain adequate insulin secretion and to maintain euglycemia for prolonged periods free of continued pharmacotherapy. Whether observations made in (mostly rodent) models of diabetes mellitus and in clinical trials are revealing identical mechanisms and therapeutic opportunities remains a tantalizing possibility. Our intention is for this review to serve as an overview of the field and commentary of this particularly exciting field of research.
Collapse
Affiliation(s)
- Wei Wang
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Chune Liu
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Maria Jimenez-Gonzalez
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Woo-Jin Song
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Mehboob A Hussain
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287.
| |
Collapse
|
26
|
Wang W, Liu C, Jimenez-Gonzalez M, Song WJ, Hussain MA. The undoing and redoing of the diabetic β-cell. J Diabetes Complications 2017; 31:912-917. [PMID: 28242267 PMCID: PMC5450161 DOI: 10.1016/j.jdiacomp.2017.01.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/23/2016] [Accepted: 01/31/2017] [Indexed: 02/08/2023]
Abstract
A hallmark of type 2 diabetes (T2DM) is the reduction in functional β-cell mass, which is considered at least in part to result from an imbalance of β-cell renewal and apoptosis, with the latter being accelerated during metabolic stress. More recent studies, however, suggest that the loss of functional β-cell mass is not as much due to β-cell death but rather to de-differentiation of β-cells when these cells are exposed to metabolic stressors, opening the possibility to re-differentiate and restore functional β-cell mass by therapeutic intervention. In parallel, clinical observations suggest that temporary intensive insulin therapy in early diagnosed humans with T2DM, so as to "rest" endogenous β-cells, allows these patients to regain adequate insulin secretion and to maintain euglycemia for prolonged periods free of continued pharmacotherapy. Whether observations made in (mostly rodent) models of diabetes mellitus and in clinical trials are revealing identical mechanisms and therapeutic opportunities remains a tantalizing possibility. Our intention is for this review to serve as an overview of the field and commentary of this particularly exciting field of research.
Collapse
Affiliation(s)
- Wei Wang
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Chune Liu
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Maria Jimenez-Gonzalez
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Woo-Jin Song
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Mehboob A Hussain
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287.
| |
Collapse
|
27
|
Hunter CS, Stein RW. Evidence for Loss in Identity, De-Differentiation, and Trans-Differentiation of Islet β-Cells in Type 2 Diabetes. Front Genet 2017; 8:35. [PMID: 28424732 PMCID: PMC5372778 DOI: 10.3389/fgene.2017.00035] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/13/2017] [Indexed: 01/31/2023] Open
Abstract
The two main types of diabetes mellitus have distinct etiologies, yet a similar outcome: loss of islet β-cell function that is solely responsible for the secretion of the insulin hormone to reduce elevated plasma glucose toward euglycemic levels. Type 1 diabetes (T1D) has traditionally been characterized by autoimmune-mediated β-cell death leading to insulin-dependence, whereas type 2 diabetes (T2D) has hallmarks of peripheral insulin resistance, β-cell dysfunction, and cell death. However, a growing body of evidence suggests that, especially during T2D, key components of β-cell failure involves: (1) loss of cell identity, specifically proteins associated with mature cell function (e.g., insulin and transcription factors like MAFA, PDX1, and NKX6.1), as well as (2) de-differentiation, defined by regression to a progenitor or stem cell-like state. New technologies have allowed the field to compare islet cell characteristics from normal human donors to those under pathophysiological conditions by single cell RNA-Sequencing and through epigenetic analysis. This has revealed a remarkable level of heterogeneity among histologically defined "insulin-positive" β-cells. These results not only suggest that these β-cell subsets have different responses to insulin secretagogues, but that defining their unique gene expression and epigenetic modification profiles will offer opportunities to develop cellular therapeutics to enrich/maintain certain subsets for correcting pathological glucose levels. In this review, we will summarize the recent literature describing how β-cell heterogeneity and plasticity may be influenced in T2D, and various possible avenues of therapeutic intervention.
Collapse
Affiliation(s)
- Chad S Hunter
- Division of Endocrinology, Diabetes and Metabolism, Comprehensive Diabetes Center and Department of Medicine, University of Alabama at BirminghamBirmingham, AL, USA
| | - Roland W Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt UniversityNashville, TN, USA
| |
Collapse
|
28
|
Reno CM, Puente EC, Sheng Z, Daphna-Iken D, Bree AJ, Routh VH, Kahn BB, Fisher SJ. Brain GLUT4 Knockout Mice Have Impaired Glucose Tolerance, Decreased Insulin Sensitivity, and Impaired Hypoglycemic Counterregulation. Diabetes 2017; 66:587-597. [PMID: 27797912 PMCID: PMC5319720 DOI: 10.2337/db16-0917] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/12/2016] [Indexed: 12/12/2022]
Abstract
GLUT4 in muscle and adipose tissue is important in maintaining glucose homeostasis. However, the role of insulin-responsive GLUT4 in the central nervous system has not been well characterized. To assess its importance, a selective knockout of brain GLUT4 (BG4KO) was generated by crossing Nestin-Cre mice with GLUT4-floxed mice. BG4KO mice had a 99% reduction in GLUT4 protein expression throughout the brain. Despite normal feeding and fasting glycemia, BG4KO mice were glucose intolerant, demonstrated hepatic insulin resistance, and had reduced glucose uptake in the brain. In response to hypoglycemia, BG4KO mice had impaired glucose sensing, noted by impaired epinephrine and glucagon responses and impaired c-fos activation in the hypothalamic paraventricular nucleus. Moreover, in vitro glucose sensing of glucose-inhibitory neurons from the ventromedial hypothalamus was impaired in BG4KO mice. In summary, BG4KO mice are glucose intolerant, insulin resistant, and have impaired glucose sensing, indicating a critical role for brain GLUT4 in sensing and responding to changes in blood glucose.
Collapse
Affiliation(s)
- Candace M Reno
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University in St. Louis, St. Louis, MO
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Erwin C Puente
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Zhenyu Sheng
- Department of Pharmacology and Physiology, Rutgers New Jersey Medical School, Newark, NJ
| | - Dorit Daphna-Iken
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Adam J Bree
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Vanessa H Routh
- Department of Pharmacology and Physiology, Rutgers New Jersey Medical School, Newark, NJ
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Simon J Fisher
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University in St. Louis, St. Louis, MO
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| |
Collapse
|
29
|
CCL5/RANTES contributes to hypothalamic insulin signaling for systemic insulin responsiveness through CCR5. Sci Rep 2016; 6:37659. [PMID: 27898058 PMCID: PMC5127185 DOI: 10.1038/srep37659] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/31/2016] [Indexed: 11/08/2022] Open
Abstract
Many neurodegenerative diseases are accompanied by metabolic disorders. CCL5/RANTES, and its receptor CCR5 are known to contribute to neuronal function as well as to metabolic disorders such as type 2 diabetes mellitus, obesity, atherosclerosis and metabolic changes after HIV infection. Herein, we found that the lack of CCR5 or CCL5 in mice impaired regulation of energy metabolism in hypothalamus. Immunostaining and co-immunoprecipitation revealed the specific expression of CCR5, associated with insulin receptors, in the hypothalamic arcuate nucleus (ARC). Both ex vivo stimulation and in vitro tissue culture studies demonstrated that the activation of insulin, and PI3K-Akt pathways were impaired in CCR5 and CCL5 deficient hypothalamus. The inhibitory phosphorylation of insulin response substrate-1 at Ser302 (IRS-1S302) but not IRS-2, by insulin was markedly increased in CCR5 and CCL5 deficient animals. Elevating CCR5/CCL5 activity induced GLUT4 membrane translocation and reduced phospho-IRS-1S302 through AMPKα-S6 Kinase. Blocking CCR5 using the antagonist, MetCCL5, abolished the de-phosphorylation of IRS-1S302 and insulin signal activation. In addition, intracerebroventricular delivery of MetCCL5 interrupted hypothalamic insulin signaling and elicited peripheral insulin responsiveness and glucose intolerance. Taken together, our data suggest that CCR5 regulates insulin signaling in hypothalamus which contributes to systemic insulin sensitivity and glucose metabolism.
Collapse
|
30
|
Li Z, Frey JL, Wong GW, Faugere MC, Wolfgang MJ, Kim JK, Riddle RC, Clemens TL. Glucose Transporter-4 Facilitates Insulin-Stimulated Glucose Uptake in Osteoblasts. Endocrinology 2016; 157:4094-4103. [PMID: 27689415 PMCID: PMC5086531 DOI: 10.1210/en.2016-1583] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent studies have identified the osteoblast as an insulin responsive cell that participates in global energy homeostasis. Here, we show that glucose transporter-4 (Glut4) is required for insulin-dependent uptake and oxidation of glucose in mature osteoblasts. In primary cultures of mouse osteoblasts, insulin increased uptake and oxidation of 14C-glucose in a dose-dependent fashion but did not significantly affect uptake or oxidation of 14C-oleate. In vitro, undifferentiated osteoblasts expressed 3 high-affinity Gluts: Glut1, Glut4, and Glut3. However, although levels of Glut1 and Glut3 remained constant during the course of osteoblast differentiation, Glut4 expression increased by 5-fold in association with enhanced insulin-stimulated glucose uptake. Glut4 ablation in osteoblasts in vitro eliminated insulin-stimulated glucose uptake, reduced proliferation and diminished measures of osteoblast maturation. In vivo, Glut4 expression was observed in osteoblasts, osteocytes, and chondrocytes at a level approaching that observed in adjacent skeletal muscle. To determine the importance of Glut4 in bone in vivo, we generated mice lacking Glut4 in osteoblasts and osteocytes (ΔGlut4). ΔGlut4 mice exhibited normal bone architecture but exhibited an increase in peripheral fat in association with hyperinsulinemia, β-cell islet hypertrophy, and reduced insulin sensitivity. Surprisingly, the expression of insulin target genes in liver, muscle, and adipose from ΔGlut4 mice were unchanged or increased, indicating that alterations in glucose homeostasis were the result of reduced clearance by bone. These findings suggest that Glut4 mediates insulin-stimulated glucose uptake by mature osteoblasts/osteocytes and that the magnitude of glucose use by bone cells is sufficient to impact global glucose disposal in the mouse.
Collapse
Affiliation(s)
- Zhu Li
- Department of Orthopaedic Surgery (Z.L., J.L.F., R.C.R., T.L.C.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Physiology (G.W.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Medicine (M.-C.F.), Division of Nephology, University of Kentucky, Lexington, Kentucky 40356; Department of Biological Chemistry (M.J.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Wooster, Massachusetts 01605; and Baltimore Veterans Administration Medical Center (R.C.R., T.L.C.), Baltimore, Maryland 21201
| | - Julie L Frey
- Department of Orthopaedic Surgery (Z.L., J.L.F., R.C.R., T.L.C.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Physiology (G.W.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Medicine (M.-C.F.), Division of Nephology, University of Kentucky, Lexington, Kentucky 40356; Department of Biological Chemistry (M.J.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Wooster, Massachusetts 01605; and Baltimore Veterans Administration Medical Center (R.C.R., T.L.C.), Baltimore, Maryland 21201
| | - G William Wong
- Department of Orthopaedic Surgery (Z.L., J.L.F., R.C.R., T.L.C.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Physiology (G.W.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Medicine (M.-C.F.), Division of Nephology, University of Kentucky, Lexington, Kentucky 40356; Department of Biological Chemistry (M.J.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Wooster, Massachusetts 01605; and Baltimore Veterans Administration Medical Center (R.C.R., T.L.C.), Baltimore, Maryland 21201
| | - Marie-Claude Faugere
- Department of Orthopaedic Surgery (Z.L., J.L.F., R.C.R., T.L.C.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Physiology (G.W.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Medicine (M.-C.F.), Division of Nephology, University of Kentucky, Lexington, Kentucky 40356; Department of Biological Chemistry (M.J.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Wooster, Massachusetts 01605; and Baltimore Veterans Administration Medical Center (R.C.R., T.L.C.), Baltimore, Maryland 21201
| | - Michael J Wolfgang
- Department of Orthopaedic Surgery (Z.L., J.L.F., R.C.R., T.L.C.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Physiology (G.W.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Medicine (M.-C.F.), Division of Nephology, University of Kentucky, Lexington, Kentucky 40356; Department of Biological Chemistry (M.J.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Wooster, Massachusetts 01605; and Baltimore Veterans Administration Medical Center (R.C.R., T.L.C.), Baltimore, Maryland 21201
| | - Jason K Kim
- Department of Orthopaedic Surgery (Z.L., J.L.F., R.C.R., T.L.C.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Physiology (G.W.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Medicine (M.-C.F.), Division of Nephology, University of Kentucky, Lexington, Kentucky 40356; Department of Biological Chemistry (M.J.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Wooster, Massachusetts 01605; and Baltimore Veterans Administration Medical Center (R.C.R., T.L.C.), Baltimore, Maryland 21201
| | - Ryan C Riddle
- Department of Orthopaedic Surgery (Z.L., J.L.F., R.C.R., T.L.C.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Physiology (G.W.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Medicine (M.-C.F.), Division of Nephology, University of Kentucky, Lexington, Kentucky 40356; Department of Biological Chemistry (M.J.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Wooster, Massachusetts 01605; and Baltimore Veterans Administration Medical Center (R.C.R., T.L.C.), Baltimore, Maryland 21201
| | - Thomas L Clemens
- Department of Orthopaedic Surgery (Z.L., J.L.F., R.C.R., T.L.C.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Physiology (G.W.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Medicine (M.-C.F.), Division of Nephology, University of Kentucky, Lexington, Kentucky 40356; Department of Biological Chemistry (M.J.W.), Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Wooster, Massachusetts 01605; and Baltimore Veterans Administration Medical Center (R.C.R., T.L.C.), Baltimore, Maryland 21201
| |
Collapse
|
31
|
Aldehyde dehydrogenase 1a3 defines a subset of failing pancreatic β cells in diabetic mice. Nat Commun 2016; 7:12631. [PMID: 27572106 PMCID: PMC5013715 DOI: 10.1038/ncomms12631] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 07/18/2016] [Indexed: 12/21/2022] Open
Abstract
Insulin-producing β cells become dedifferentiated during diabetes progression. An impaired ability to select substrates for oxidative phosphorylation, or metabolic inflexibility, initiates progression from β-cell dysfunction to β-cell dedifferentiation. The identification of pathways involved in dedifferentiation may provide clues to its reversal. Here we isolate and functionally characterize failing β cells from various experimental models of diabetes and report a striking enrichment in the expression of aldehyde dehydrogenase 1 isoform A3 (ALDH(+)) as β cells become dedifferentiated. Flow-sorted ALDH(+) islet cells demonstrate impaired glucose-induced insulin secretion, are depleted of Foxo1 and MafA, and include a Neurogenin3-positive subset. RNA sequencing analysis demonstrates that ALDH(+) cells are characterized by: (i) impaired oxidative phosphorylation and mitochondrial complex I, IV and V; (ii) activated RICTOR; and (iii) progenitor cell markers. We propose that impaired mitochondrial function marks the progression from metabolic inflexibility to dedifferentiation in the natural history of β-cell failure.
Collapse
|
32
|
Zhou PZ, Zhu YM, Zou GH, Sun YX, Xiu XL, Huang X, Zhang QH. Relationship Between Glucocorticoids and Insulin Resistance in Healthy Individuals. Med Sci Monit 2016; 22:1887-94. [PMID: 27258456 PMCID: PMC4913831 DOI: 10.12659/msm.895251] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background The aim of this study was to determine the correlation between glucocorticoids (GCs) and insulin resistance (IR) in healthy individuals by conducting a systematic meta-analysis. Material/Methods A systematic literature review was conducted using 9 electronic databases. Only case-control studies investigating fasting plasma glucose (FPG) and IR were enrolled based on strictly established selection criteria. Statistical analyses were performed by Stata software, version 12.0 (Stata Corporation, College Station, Texas, USA). Results Among 496 initially retrieved articles, only 6 papers published in English were eventually included in this meta-analysis. A total of 201 healthy individuals (105 in GC group and 96 in control group) were included in the 6 studies. In 4 of these 6 studies, dexamethasone was used, and in the other 2 studies prednisolone was given. This meta-analysis revealed that the FPG, fasting insulin (FINS), and homeostasis model assessment of insulin resistance (HOMA-IR) levels in the GC group were all significantly higher than that in the control group (FPG: SMD=2.65, 95%CI=1.42~3.88, P<0.001; FINS: SMD=2.48, 95%CI=1.01~3.95, P=0.001; HOMA-IR: SMD=38.30, 95%CI=24.38~52.22, P<0.001). Conclusions In conclusion, our present study revealed that therapies using GCs might result in elevated FPG, FINS, and HOMA-IR, and thereby contribute to IR in healthy individuals.
Collapse
Affiliation(s)
- Peng-Zhen Zhou
- Department of Reproductive Medicine, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China (mainland)
| | - Yong-Mei Zhu
- Department of Pediatric Orthopaedic, Yantaishan Hospital, Yantai, Shandong, China (mainland)
| | - Guang-Hui Zou
- Department of Reproductive Medicine, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China (mainland)
| | - Yu-Xia Sun
- Department of Reproductive Medicine, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China (mainland)
| | - Xiao-Lin Xiu
- Department of Reproductive Medicine, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China (mainland)
| | - Xin Huang
- Department of Reproductive Medicine, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China (mainland)
| | - Qun-Hui Zhang
- Department of Rheumatism and Immunology, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China (mainland)
| |
Collapse
|
33
|
Abstract
Until recently, type 2 diabetes was seen as a disease caused by an impaired ability of insulin to promote the uptake and utilisation of glucose. Work on forkhead box protein O (FOXO) transcription factors revealed new aspects of insulin action that have led us to articulate a liver- and beta cell-centric narrative of diabetes pathophysiology and treatment. FOXO integrate a surprisingly diverse subset of biological functions to promote metabolic flexibility. In the liver, they controls the glucokinase/glucose-6-phosphatase switch and bile acid pool composition, directing carbons to glucose or lipid utilisation, thus providing a unifying mechanism for the two abnormalities of the diabetic liver: excessive glucose production and increased lipid synthesis and secretion. Moreover, FOXO are necessary to maintain beta cell differentiation, and diabetes development is associated with a gradual loss of FOXO function that brings about beta cell dedifferentiation. We proposed that dedifferentiation is the main cause of beta cell failure and conversion into non-beta endocrine cells, and that treatment should restore beta cell differentiation. Our studies investigating these proposals have revealed new dimensions to the pathophysiology of diabetes that can be leveraged to design new therapies.
Collapse
Affiliation(s)
- Utpal B Pajvani
- Department of Medicine and Naomi Berrie Diabetes Center, Columbia University Medical Center, 1150 St Nicholas Av., New York, NY, 10032, USA.
| | - Domenico Accili
- Department of Medicine and Naomi Berrie Diabetes Center, Columbia University Medical Center, 1150 St Nicholas Av., New York, NY, 10032, USA.
| |
Collapse
|
34
|
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: 91] [Impact Index Per Article: 9.1] [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
|
35
|
Ren H, Lu TY, McGraw TE, Accili D. Anorexia and impaired glucose metabolism in mice with hypothalamic ablation of Glut4 neurons. Diabetes 2015; 64:405-17. [PMID: 25187366 PMCID: PMC4303970 DOI: 10.2337/db14-0752] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The central nervous system (CNS) uses glucose independent of insulin. Nonetheless, insulin receptors and insulin-responsive glucose transporters (Glut4) often colocalize in neurons (Glut4 neurons) in anatomically and functionally distinct areas of the CNS. The apparent heterogeneity of Glut4 neurons has thus far thwarted attempts to understand their function. To answer this question, we used Cre-dependent, diphtheria toxin-mediated cell ablation to selectively remove basal hypothalamic Glut4 neurons and investigate the resulting phenotypes. After Glut4 neuron ablation, mice demonstrate altered hormone and nutrient signaling in the CNS. Accordingly, they exhibit negative energy balance phenotype characterized by reduced food intake and increased energy expenditure, without locomotor deficits or gross neuronal abnormalities. Glut4 neuron ablation affects orexigenic melanin-concentrating hormone neurons but has limited effect on neuropeptide Y/agouti-related protein and proopiomelanocortin neurons. The food intake phenotype can be partially normalized by GABA administration, suggesting that it arises from defective GABAergic transmission. Glut4 neuron-ablated mice show peripheral metabolic defects, including fasting hyperglycemia and glucose intolerance, decreased insulin levels, and elevated hepatic gluconeogenic genes. We conclude that Glut4 neurons integrate hormonal and nutritional cues and mediate CNS actions of insulin on energy balance and peripheral metabolism.
Collapse
Affiliation(s)
- Hongxia Ren
- Department of Medicine and Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, NY
| | - Taylor Y Lu
- Department of Medicine and Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, NY
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medical College, New York, NY
| | - Domenico Accili
- Department of Medicine and Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, NY
| |
Collapse
|
36
|
Ren H, Yan S, Zhang B, Lu TY, Arancio O, Accili D. Glut4 expression defines an insulin-sensitive hypothalamic neuronal population. Mol Metab 2014; 3:452-9. [PMID: 24944904 PMCID: PMC4060214 DOI: 10.1016/j.molmet.2014.04.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 04/10/2014] [Indexed: 11/19/2022] Open
Abstract
Insulin signaling in the CNS modulates satiety and glucose metabolism, but insulin target neurons are poorly defined. We have previously shown that ablation of insulin receptors (InsR) in Glut4-expressing tissues results in systemic abnormalities of insulin action. We propose that Glut4 neurons constitute an insulin-sensitive neuronal subset. We determined their gene expression profiles using flow-sorted hypothalamic Glut4 neurons. Gene ontology analyses demonstrated that Glut4 neurons are enriched in olfacto-sensory receptors, M2 acetylcholine receptors, and pathways required for the acquisition of insulin sensitivity. Following genetic ablation of InsR, transcriptome profiling of Glut4 neurons demonstrated impairment of the insulin, peptide hormone, and cAMP signaling pathways, with a striking upregulation of anion homeostasis pathway. Accordingly, hypothalamic InsR-deficient Glut4 neurons showed reduced firing activity. The molecular signature of Glut4 neurons is consistent with a role for this neural population in the integration of olfacto-sensory cues with hormone signaling to regulate peripheral metabolism.
Collapse
Affiliation(s)
- Hongxia Ren
- Department of Medicine and Berrie Diabetes Center, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA
| | - Shijun Yan
- Department of Pathology and Cell Biology, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA
| | - Baifang Zhang
- Department of Medicine and Berrie Diabetes Center, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA
| | - Taylor Y. Lu
- Department of Medicine and Berrie Diabetes Center, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA
| | - Domenico Accili
- Department of Medicine and Berrie Diabetes Center, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA
- Corresponding author.
| |
Collapse
|
37
|
Cloning and functional analysis of goat glucose transporter 4. Mol Biol Rep 2014; 41:757-68. [PMID: 24381100 DOI: 10.1007/s11033-013-2915-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 12/17/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Glucose transporter (GLUT) 4 is a major mediator of blood glucose levels and a key regulator of whole-body glucose homeostasis. This study aimed at evaluating the function of goat GLUT4 on glucose absorption and the effect of GLUT4 on lactose synthesis in goat mammary gland epithelial (GMGE) cells. METHODS Currently, the cDNA of GLUT4, a putative facilitative GLUT, was cloned from goat. To investigate the function of goat GLUT4, we constructed the eukaryotic expression vector pcDNA3.1-GLUT4 and used it to transfect GMGE cells, and then GLUT4 transfected GMGE (G4T-GMGE) cells were generated. The deduced GLUT4 sequence comprised 509 amino acids, what meant that a putative protein with a molecular weight of approximately 55 kDa would be produced. Both glucose uptake and lactose synthesis increased in the G4T-GMGE cells compared with the GMGE cells. At the transcriptional level, GLUT4 expression increased by nearly 55-fold in the G4T-GMGE cells, and the expression of amino acid transporters (SLC1A5, SLC3A2 and SLC7A5) enhanced as well; in contrast, GLUT1 expression decreased by more than 50 % in the G4T-GMGE cells. CONCLUSION These results suggest that goat GLUT4 functions in the transport of glucose and it may play a positive role in amino acid uptake in mammary glands.
Collapse
|
38
|
Cherng YG, Tsai CC, Chung HH, Lai YW, Kuo SC, Cheng JT. Antihyperglycemic action of sinapic acid in diabetic rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2013; 61:12053-9. [PMID: 24261449 DOI: 10.1021/jf403092b] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Sinapic acid is a hydroxycinnamic acid contained in plants. In an attempt to know the hyperglycemic effect of sinapic acid, this study applied streptozotocin (STZ) to induce type 1-like diabetic rats and fed fructose-rich chow to induce type 2-like diabetic rats. Sinapic acid dose-dependently reduced the hyperglycemia of STZ-diabetic rats (9.8 ± 1.8%, 11.6 ± 0.7%, and 19.4 ± 3.2% at 5 mg/kg, 10 mg/kg, and 25 mg/kg, respectively). Also, sinapic acid attenuated the postprandial plasma glucose without changing plasma insulin in rats. Repeated treatment of sinapic acid increased the gene expression of GLUT4 in soleus muscle of STZ-diabetic rats. Moreover, sinapic acid enhanced glucose uptake into isolated soleus muscle and L6 cells (337.0 ± 29.6%). Inhibition of phospholipase C (PLC) using U73122 (1.00 ± 0.02 μg/mg protein) or protein kinase C (PKC) using chelerythrine (0.97 ± 0.02 μg/mg protein) attenuated the sinapic acid-stimulated glucose uptake (1.63 ± 0.02 μg/mg protein) in L6 cells. Otherwise, the reduced glucose infusion rate (GIR) in fructose-rich chow-fed rats was also raised by sinapic acid. Our results suggest that sinapic acid ameliorates hyperglycemia through PLC-PKC signals to enhance the glucose utilization in diabetic rats.
Collapse
Affiliation(s)
- Yih-Giun Cherng
- Department of Anesthesiology, Shuang Ho Hospital , and Department of Anesthesiology, College of Medicine, Taipei Medical University , New Taipei City 10361, Taiwan
| | | | | | | | | | | |
Collapse
|
39
|
Ramalingam L, Oh E, Thurmond DC. Novel roles for insulin receptor (IR) in adipocytes and skeletal muscle cells via new and unexpected substrates. Cell Mol Life Sci 2013; 70:2815-34. [PMID: 23052216 PMCID: PMC3556358 DOI: 10.1007/s00018-012-1176-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 08/21/2012] [Accepted: 09/18/2012] [Indexed: 01/30/2023]
Abstract
The insulin signaling pathway regulates whole-body glucose homeostasis by transducing extracellular signals from the insulin receptor (IR) to downstream intracellular targets, thus coordinating a multitude of biological functions. Dysregulation of IR or its signal transduction is associated with insulin resistance, which may culminate in type 2 diabetes. Following initial stimulation of IR, insulin signaling diverges into different pathways, activating multiple substrates that have roles in various metabolic and cellular processes. The integration of multiple pathways arising from IR activation continues to expand as new IR substrates are identified and characterized. Accordingly, our review will focus on roles for IR substrates as they pertain to three primary areas: metabolism/glucose uptake, mitogenesis/growth, and aging/longevity. While IR functions in a seemingly pleiotropic manner in many cell types, through these three main roles in fat and skeletal muscle cells, IR multi-tasks to regulate whole-body glucose homeostasis to impact healthspan and lifespan.
Collapse
Affiliation(s)
- Latha Ramalingam
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Eunjin Oh
- Department of Pediatrics, Herman B Wells Center, Indiana University School of Medicine, Indianapolis, IN USA
| | - Debbie C. Thurmond
- Departments of Pediatrics, Biochemistry and Molecular Biology, and Cellular and Integrative Physiology, Herman B Wells Center, Indiana University School of Medicine, 635 Barnhill Drive MS 2031, Indianapolis, IN 46202 USA
| |
Collapse
|
40
|
Abstract
Diabetes affects more than 300 million individuals globally, contributing to significant morbidity and mortality worldwide. As the incidence and prevalence of diabetes continue to escalate with the force of an approaching tsunami, it is imperative that we better define the biological mechanisms causing both obesity and diabetes and identify optimal prevention and treatment strategies that will enable a healthier environment and calmer waters. New guidelines from the American Diabetes Association/European Association of the Study of Diabetes and The Endocrine Society encourage individualized care for each patient with diabetes, both in the outpatient and inpatient setting. Recent data suggest that restoration of normal glucose metabolism in people with prediabetes may delay progression to type 2 diabetes (T2DM). However, several large clinical trials have underscored the limitations of current treatment options once T2DM has developed, particularly in obese children with the disease. Prospects for reversing new-onset type 1 diabetes also appear limited, although recent clinical trials indicate that immunotherapy can delay the loss of β-cell function, suggesting potential benefits if treatment is initiated earlier. Research demonstrating a role for the central nervous system in the development of obesity and T2DM, the identification of a new hormone that simulates some of the benefits of exercise, and the development of new β-cell imaging techniques may provide novel therapeutic targets and biomarkers of early diabetes detection for optimization of interventions. Today's message is that a diabetes tsunami is imminent, and the only way to minimize the damage is to create an early warning system and improve interventions to protect those in its path.
Collapse
Affiliation(s)
- R Sherwin
- Department of Internal Medicine, Division of Endocrinology, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| | | |
Collapse
|
41
|
Talchai C, Xuan S, Lin HV, Sussel L, Accili D. Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure. Cell 2012; 150:1223-34. [PMID: 22980982 DOI: 10.1016/j.cell.2012.07.029] [Citation(s) in RCA: 1098] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 03/20/2012] [Accepted: 07/06/2012] [Indexed: 12/12/2022]
Abstract
Diabetes is associated with β cell failure. But it remains unclear whether the latter results from reduced β cell number or function. FoxO1 integrates β cell proliferation with adaptive β cell function. We interrogated the contribution of these two processes to β cell dysfunction, using mice lacking FoxO1 in β cells. FoxO1 ablation caused hyperglycemia with reduced β cell mass following physiologic stress, such as multiparity and aging. Surprisingly, lineage-tracing experiments demonstrated that loss of β cell mass was due to β cell dedifferentiation, not death. Dedifferentiated β cells reverted to progenitor-like cells expressing Neurogenin3, Oct4, Nanog, and L-Myc. A subset of FoxO1-deficient β cells adopted the α cell fate, resulting in hyperglucagonemia. Strikingly, we identify the same sequence of events as a feature of different models of murine diabetes. We propose that dedifferentiation trumps endocrine cell death in the natural history of β cell failure and suggest that treatment of β cell dysfunction should restore differentiation, rather than promoting β cell replication.
Collapse
Affiliation(s)
- Chutima Talchai
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
42
|
Ren W, Cheema S, Du K. The association of ClipR-59 protein with AS160 modulates AS160 protein phosphorylation and adipocyte Glut4 protein membrane translocation. J Biol Chem 2012; 287:26890-900. [PMID: 22689584 DOI: 10.1074/jbc.m112.357699] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
ClipR-59 is a membrane-associated protein and has been implicated in membrane signaling and vesicle trafficking. Recently, we have identified ClipR-59 as an Akt-interacting protein, and we have found that, by interacting with Akt, ClipR-59 modulates Akt subcellular compartmentalization and Akt substrate AS160 phosphorylation, thereby promoting Glut4 membrane translocation. Here, we have further investigated the regulatory effects of ClipR-59 on AS160 phosphorylation and subsequent adipocyte glucose transport. Our data showed that ClipR-59 interacted with AS160, which was mediated by the ankyrin repeats of ClipR-59 and regulated by insulin signaling. Moreover, the data also demonstrated that the interaction of ClipR-59 with AS160 was required for ClipR-59 to modulate Glut4 membrane translocation as ΔANK-ClipR-59, an AS160 interaction-defective mutant, failed to promote AS160 phosphorylation, Glut4 membrane translocation, and glucose transport induced by insulin in 3T3-L1 adipocytes. Because ClipR-59 also interacts with Akt and enhances the interaction between Akt and AS160, we suggest that ClipR-59 functions as a scaffold protein to facilitate Akt-mediated AS160 phosphorylation, thereby regulating glucose transport.
Collapse
Affiliation(s)
- Wenying Ren
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts 02111, USA
| | | | | |
Collapse
|
43
|
Si Y, Zhao Y, Hao H, Liu J, Guo Y, Mu Y, Shen J, Cheng Y, Fu X, Han W. Infusion of mesenchymal stem cells ameliorates hyperglycemia in type 2 diabetic rats: identification of a novel role in improving insulin sensitivity. Diabetes 2012; 61:1616-25. [PMID: 22618776 PMCID: PMC3357293 DOI: 10.2337/db11-1141] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Infusion of mesenchymal stem cells (MSCs) has been shown to effectively lower blood glucose in diabetic individuals, but the mechanism involved could not be adequately explained by their potential role in promoting islet regeneration. We therefore hypothesized that infused MSCs might also contribute to amelioration of the insulin resistance of peripheral insulin target tissues. To test the hypothesis, we induced a diabetic rat model by high-fat diet/streptozotocin (STZ) administration, performed MSC infusion during the early phase (7 days) or late phase (21 days) after STZ injection, and then evaluated the therapeutic effects of MSC infusion and explored the possible mechanisms involved. MSC infusion ameliorated hyperglycemia in rats with type 2 diabetes (T2D). Infusion of MSCs during the early phase not only promoted β-cell function but also ameliorated insulin resistance, whereas infusion in the late phase merely ameliorated insulin resistance. Infusion of MSCs resulted in an increase of GLUT4 expression and an elevation of phosphorylated insulin receptor substrate 1 (IRS-1) and Akt (protein kinase B) in insulin target tissues. This is the first report of MSC treatment improving insulin sensitivity in T2D. These data indicate that multiple roles and mechanisms are involved in the efficacy of MSCs in ameliorating hyperglycemia in T2D.
Collapse
Affiliation(s)
- Yiling Si
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Yali Zhao
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Haojie Hao
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Jiejie Liu
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Yelei Guo
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Yiming Mu
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, China
| | - Jing Shen
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, China
| | - Yu Cheng
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, China
| | - Xiaobing Fu
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
- Corresponding author: Weidong Han, , or Xiaobing Fu,
| | - Weidong Han
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
- Corresponding author: Weidong Han, , or Xiaobing Fu,
| |
Collapse
|
44
|
Ramnanan CJ, Edgerton DS, Cherrington AD. Evidence against a physiologic role for acute changes in CNS insulin action in the rapid regulation of hepatic glucose production. Cell Metab 2012; 15:656-64. [PMID: 22560218 PMCID: PMC3348512 DOI: 10.1016/j.cmet.2012.03.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This Perspective will discuss the physiologic relevance of data that suggest CNS insulin action is required for the rapid suppression of hepatic glucose production. It will also review data from experiments on the conscious dog, which show that although the canine brain can sense insulin and, thereby, regulate hepatic glucoregulatory enzyme expression, CNS insulin action is not essential for the rapid suppression of glucose production caused by the hormone. Insulin's direct hepatic effects are dominant, thus it appears that insulin's central effects are redundant in the acute regulation of hepatic glucose metabolism.
Collapse
Affiliation(s)
- Christopher J Ramnanan
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, TN 37232, USA
| | | | | |
Collapse
|
45
|
Affiliation(s)
- Owen Chan
- Department of Internal Medicine, Section of Endocrinology, Yale School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
46
|
Paranjape SA, Chan O, Zhu W, Horblitt AM, Grillo CA, Wilson S, Reagan L, Sherwin RS. Chronic reduction of insulin receptors in the ventromedial hypothalamus produces glucose intolerance and islet dysfunction in the absence of weight gain. Am J Physiol Endocrinol Metab 2011; 301:E978-83. [PMID: 21828334 PMCID: PMC3774165 DOI: 10.1152/ajpendo.00304.2011] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Insulin is believed to regulate glucose homeostasis mainly via direct effects on the liver, muscle, and adipose tissues. The contribution of insulin's central nervous system effects to disorders of glucose metabolism has received less attention. To evaluate whether postnatal reduction of insulin receptors (IRs) within the ventromedial hypothalamus (VMH), a brain region critical for glucose sensing, contributes to disorders of peripheral glucose metabolism, we microinjected a lentiviral vector expressing an antisense sequence to knockdown IRs or a control lentiviral vector into the VMH of nonobese nondiabetic rats. After 3-4 mo, we assessed 1) glucose tolerance, 2) hepatic insulin sensitivity, and 3) insulin and glucagon secretion, using the glucose clamp technique. Knockdown of IRs locally in the VMH caused glucose intolerance without altering body weight. Increments of plasma insulin during a euglycemic clamp study failed to suppress endogenous glucose production and produced a paradoxical rise in plasma glucagon in the VMH-IR knockdown rats. Unexpectedly, these animals also displayed a 40% reduction (P < 0.05) in insulin secretion in response to an identical hyperglycemic stimulus (∼220 mg/dl). Our data demonstrate that chronic suppression of VMH-IR gene expression is sufficient to impair glucose metabolism as well as α-cell and β-cell function in nondiabetic, nonobese rats. These data suggest that insulin resistance within the VMH may be a significant contributor to the development of type 2 diabetes.
Collapse
Affiliation(s)
- Sachin A Paranjape
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Levin BE, Sherwin RS. Peripheral glucose homeostasis: does brain insulin matter? J Clin Invest 2011; 121:3392-5. [PMID: 21865650 DOI: 10.1172/jci59653] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Much controversy surrounds the relative role of insulin signaling in the brain in the control of hepatic glucose metabolism. In this issue of the JCI, Ramnanan and colleagues demonstrate that arterial infusion of insulin into the brains of dogs reduces net hepatic glucose output without altering endogenous glucose production. However, this effect was modest and required both prolonged fasting and prolonged exposure of the brain to insulin, raising doubts about the overall physiological relevance of insulin action in the brain on hepatic glucose metabolism. Given the dominant direct role that insulin plays in inhibiting glucose production in the liver, we suggest that the main effect of central insulin on hepatic glucose metabolism may be more chronic and assume greater significance either when portal insulin is deficient, as occurs during exogenous insulin treatment of type 1 diabetes, or when chronic hyperinsulinemia and central insulin resistance develops, as occurs in type 2 diabetes.
Collapse
Affiliation(s)
- Barry E Levin
- Neurology Service (127C), VA Medical Center, East Orange, New Jersey 07018, USA.
| | | |
Collapse
|
48
|
Lin HV, Accili D. Reconstitution of insulin action in muscle, white adipose tissue, and brain of insulin receptor knock-out mice fails to rescue diabetes. J Biol Chem 2011; 286:9797-804. [PMID: 21239487 DOI: 10.1074/jbc.m110.210807] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Type 2 diabetes results from an impairment of insulin action. The first demonstrable abnormality of insulin signaling is a decrease of insulin-dependent glucose disposal followed by an increase in hepatic glucose production. In an attempt to dissect the relative importance of these two changes in disease progression, we have employed genetic knock-outs/knock-ins of the insulin receptor. Previously, we demonstrated that insulin receptor knock-out mice (Insr(-/-)) could be rescued from diabetes by reconstitution of insulin signaling in liver, brain, and pancreatic β cells (L1 mice). In this study, we used a similar approach to reconstitute insulin signaling in tissues that display insulin-dependent glucose uptake. Using GLUT4-Cre mice, we restored InsR expression in muscle, fat, and brain of Insr(-/-) mice (GIRKI (Glut4-insulin receptor knock-in line 1) mice). Unlike L1 mice, GIRKI mice failed to thrive and developed diabetes, although their survival was modestly extended when compared with Insr(-/-). The data underscore the role of developmental factors in the presentation of murine diabetes. The broader implication of our findings is that diabetes treatment should not necessarily target the same tissues that are responsible for disease pathogenesis.
Collapse
Affiliation(s)
- Hua V Lin
- Department of Medicine, Columbia University, New York, New York 10032, USA
| | | |
Collapse
|