1
|
Rönn T, Ofori JK, Perfilyev A, Hamilton A, Pircs K, Eichelmann F, Garcia-Calzon S, Karagiannopoulos A, Stenlund H, Wendt A, Volkov P, Schulze MB, Mulder H, Eliasson L, Ruhrmann S, Bacos K, Ling C. Genes with epigenetic alterations in human pancreatic islets impact mitochondrial function, insulin secretion, and type 2 diabetes. Nat Commun 2023; 14:8040. [PMID: 38086799 PMCID: PMC10716521 DOI: 10.1038/s41467-023-43719-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Epigenetic dysregulation may influence disease progression. Here we explore whether epigenetic alterations in human pancreatic islets impact insulin secretion and type 2 diabetes (T2D). In islets, 5,584 DNA methylation sites exhibit alterations in T2D cases versus controls and are associated with HbA1c in individuals not diagnosed with T2D. T2D-associated methylation changes are found in enhancers and regions bound by β-cell-specific transcription factors and associated with reduced expression of e.g. CABLES1, FOXP1, GABRA2, GLR1A, RHOT1, and TBC1D4. We find RHOT1 (MIRO1) to be a key regulator of insulin secretion in human islets. Rhot1-deficiency in β-cells leads to reduced insulin secretion, ATP/ADP ratio, mitochondrial mass, Ca2+, and respiration. Regulators of mitochondrial dynamics and metabolites, including L-proline, glycine, GABA, and carnitines, are altered in Rhot1-deficient β-cells. Islets from diabetic GK rats present Rhot1-deficiency. Finally, RHOT1methylation in blood is associated with future T2D. Together, individuals with T2D exhibit epigenetic alterations linked to mitochondrial dysfunction in pancreatic islets.
Collapse
Affiliation(s)
- Tina Rönn
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Jones K Ofori
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Alexander Perfilyev
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Alexander Hamilton
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
- Department of Biology, University of Copenhagen, København, Denmark
| | - Karolina Pircs
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
- HCEMM-Su, Neurobiology and Neurodegenerative Diseases Research Group, Budapest, Hungary
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Fabian Eichelmann
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Sonia Garcia-Calzon
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
- Department of Food Science and Physiology, Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Alexandros Karagiannopoulos
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Hans Stenlund
- Swedish Metabolomics Centre, Umeå University, Umeå, Sweden
| | - Anna Wendt
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Petr Volkov
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Hindrik Mulder
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Lena Eliasson
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Sabrina Ruhrmann
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Karl Bacos
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Charlotte Ling
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden.
| |
Collapse
|
2
|
Zou X, Hu M, Huang X, Zhou L, Li M, Chen J, Ma L, Gao X, Luo Y, Cai X, Li Y, Zhou X, Li N, Shi Y, Han X, Ji L. Rare Variant in Metallothionein 1E Increases the Risk of Type 2 Diabetes in a Chinese Population. Diabetes Care 2023; 46:2249-2257. [PMID: 37878528 DOI: 10.2337/dc22-2031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 09/18/2023] [Indexed: 10/27/2023]
Abstract
OBJECTIVE To uncover novel targets for the treatment of type 2 diabetes (T2D) by investigating rare variants with large effects in monogenic forms of the disease. RESEARCH DESIGN AND METHODS We performed whole-exome sequencing in a family with diabetes. We validated the identified gene using Sanger sequencing in additional families and diabetes- and community-based cohorts. Wild-type and variant gene transgenic mouse models were used to study the gene function. RESULTS Our analysis revealed a rare variant of the metallothionein 1E (MT1E) gene, p.C36Y, in a three-generation family with diabetes. This risk allele was associated with T2D or prediabetes in a community-based cohort. MT1E p.C36 carriers had higher HbA1c levels and greater BMI than those carrying the wild-type allele. Mice with forced expression of MT1E p.C36Y demonstrated increased weight gain, elevated postchallenge serum glucose and liver enzyme levels, and hepatic steatosis, similar to the phenotypes observed in human carriers of MT1E p.C36Y. In contrast, mice with forced expression of MT1E p.C36C displayed reduced weight and lower serum glucose and serum triglyceride levels. Forced expression of wild-type and variant MT1E demonstrated differential expression of genes related to lipid metabolism. CONCLUSIONS Our results suggest that MT1E could be a promising target for drug development, because forced expression of MT1E p.C36C stabilized glucose metabolism and reduced body weight, whereas MT1E p.C36Y expression had the opposite effect. These findings highlight the importance of considering the impact of rare variants in the development of new T2D treatments.
Collapse
Affiliation(s)
- Xiantong Zou
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Mengdie Hu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Xiuting Huang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Lingli Zhou
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Meng Li
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Jing Chen
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Liping Ma
- Central Laboratory, Peking University People's Hospital, Beijing, China
| | - Xueying Gao
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Yingying Luo
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Yufeng Li
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
- Department of Endocrinology, Beijing Friendship Hospital Pinggu Campus, Capital Medical University, Beijing, China
| | - Xianghai Zhou
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Na Li
- Central Laboratory, Peking University People's Hospital, Beijing, China
| | - Yuanping Shi
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Xueyao Han
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, China
| |
Collapse
|
3
|
Effects of Antioxidant Gene Overexpression on Stress Resistance and Malignization In Vitro and In Vivo: A Review. Antioxidants (Basel) 2022; 11:antiox11122316. [PMID: 36552527 PMCID: PMC9774954 DOI: 10.3390/antiox11122316] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Reactive oxygen species (ROS) are normal products of a number of biochemical reactions and are important signaling molecules. However, at the same time, they are toxic to cells and have to be strictly regulated by their antioxidant systems. The etiology and pathogenesis of many diseases are associated with increased ROS levels, and many external stress factors directly or indirectly cause oxidative stress in cells. Within this context, the overexpression of genes encoding the proteins in antioxidant systems seems to have become a viable approach to decrease the oxidative stress caused by pathological conditions and to increase cellular stress resistance. However, such manipulations unavoidably lead to side effects, the most dangerous of which is an increased probability of healthy tissue malignization or increased tumor aggression. The aims of the present review were to collect and systematize the results of studies devoted to the effects resulting from the overexpression of antioxidant system genes on stress resistance and carcinogenesis in vitro and in vivo. In most cases, the overexpression of these genes was shown to increase cell and organism resistances to factors that induce oxidative and genotoxic stress but to also have different effects on cancer initiation and promotion. The last fact greatly limits perspectives of such manipulations in practice. The overexpression of GPX3 and SOD3 encoding secreted proteins seems to be the "safest" among the genes that can increase cell resistance to oxidative stress. High efficiency and safety potential can also be found for SOD2 overexpression in combinations with GPX1 or CAT and for similar combinations that lead to no significant changes in H2O2 levels. Accumulation, systematization, and the integral analysis of data on antioxidant gene overexpression effects can help to develop approaches for practical uses in biomedical and agricultural areas. Additionally, a number of factors such as genetic and functional context, cell and tissue type, differences in the function of transcripts of one and the same gene, regulatory interactions, and additional functions should be taken into account.
Collapse
|
4
|
Wang RR, Qiu X, Pan R, Fu H, Zhang Z, Wang Q, Chen H, Wu QQ, Pan X, Zhou Y, Shan P, Wang S, Guo G, Zheng M, Zhu L, Meng ZX. Dietary intervention preserves β cell function in mice through CTCF-mediated transcriptional reprogramming. J Exp Med 2022; 219:213256. [PMID: 35652891 DOI: 10.1084/jem.20211779] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 04/04/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022] Open
Abstract
Pancreatic β cell plasticity is the primary determinant of disease progression and remission of type 2 diabetes (T2D). However, the dynamic nature of β cell adaptation remains elusive. Here, we establish a mouse model exhibiting the compensation-to-decompensation adaptation of β cell function in response to increasing duration of high-fat diet (HFD) feeding. Comprehensive islet functional and transcriptome analyses reveal a dynamic orchestration of transcriptional networks featuring temporal alteration of chromatin remodeling. Interestingly, prediabetic dietary intervention completely rescues β cell dysfunction, accompanied by a remarkable reversal of HFD-induced reprogramming of islet chromatin accessibility and transcriptome. Mechanistically, ATAC-based motif analysis identifies CTCF as the top candidate driving dietary intervention-induced preservation of β cell function. CTCF expression is markedly decreased in β cells from obese and diabetic mice and humans. Both dietary intervention and AAV-mediated restoration of CTCF expression ameliorate β cell dysfunction ex vivo and in vivo, through transducing the lipid toxicity and inflammatory signals to transcriptional reprogramming of genes critical for β cell glucose metabolism and stress response.
Collapse
Affiliation(s)
- Ruo-Ran Wang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinyuan Qiu
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Ran Pan
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongxing Fu
- Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ziyin Zhang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qintao Wang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haide Chen
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qing-Qian Wu
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaowen Pan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanping Zhou
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Pengfei Shan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shusen Wang
- Organ Transplant Center, Tianjin First Central Hospital, Tianjin, China.,NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lingyun Zhu
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Geriatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
The Oxidative Balance Orchestrates the Main Keystones of the Functional Activity of Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7714542. [PMID: 35047109 PMCID: PMC8763515 DOI: 10.1155/2022/7714542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/03/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
This review is aimed at providing an overview of the key hallmarks of cardiomyocytes in physiological and pathological conditions. The main feature of cardiac tissue is the force generation through contraction. This process requires a conspicuous energy demand and therefore an active metabolism. The cardiac tissue is rich of mitochondria, the powerhouses in cells. These organelles, producing ATP, are also the main sources of ROS whose altered handling can cause their accumulation and therefore triggers detrimental effects on mitochondria themselves and other cell components thus leading to apoptosis and cardiac diseases. This review highlights the metabolic aspects of cardiomyocytes and wanders through the main systems of these cells: (a) the unique structural organization (such as different protein complexes represented by contractile, regulatory, and structural proteins); (b) the homeostasis of intracellular Ca2+ that represents a crucial ion for cardiac functions and E-C coupling; and (c) the balance of Zn2+, an ion with a crucial impact on the cardiovascular system. Although each system seems to be independent and finely controlled, the contractile proteins, intracellular Ca2+ homeostasis, and intracellular Zn2+ signals are strongly linked to each other by the intracellular ROS management in a fascinating way to form a "functional tetrad" which ensures the proper functioning of the myocardium. Nevertheless, if ROS balance is not properly handled, one or more of these components could be altered resulting in deleterious effects leading to an unbalance of this "tetrad" and promoting cardiovascular diseases. In conclusion, this "functional tetrad" is proposed as a complex network that communicates continuously in the cardiomyocytes and can drive the switch from physiological to pathological conditions in the heart.
Collapse
|
6
|
Walker S, Appari M, Forbes S. Considerations and challenges of islet transplantation and future therapies on the horizon. Am J Physiol Endocrinol Metab 2022; 322:E109-E117. [PMID: 34927459 DOI: 10.1152/ajpendo.00310.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Islet transplantation is a treatment for selected adults with type 1 diabetes and severe hypoglycemia. Islets from two or more donor pancreases, a scarce resource, are usually required to impact glycemic control, but the treatment falls short of a cure. Islets are avascular when transplanted into the hypoxic liver environment and subjected to inflammatory insults, immune attack, and toxicity from systemic immunosuppression. The Collaborative Islet Transplant Registry, with outcome data on over 1,000 islet transplant recipients, has demonstrated that larger islet numbers transplanted and older age of recipients are associated with better outcomes. Induction with T-cell depleting agents and the TNF-α inhibitor etanercept and maintenance systemic immunosuppression with mTOR inhibitors in combination with calcineurin inhibitors also appear advantageous, but concerns remain over immunosuppressive toxicity. We discuss strategies and therapeutics that address specific challenges of islet transplantation, many of which are at the preclinical stage of development. On the horizon are adjuvant cell therapies with mesenchymal stromal cells and regulatory T cells that have been used in preclinical models and in humans in other contexts; such a strategy may enable reductions in immunosuppression in the early peri-transplant period when the islets are vulnerable to apoptosis. Human embryonic stem cell-derived islets are in early-phase clinical trials and hold the promise of an inexhaustible supply of insulin-producing cells; effective encapsulation of such cells or, silencing of the human leukocyte antigen (HLA) complex would eliminate the need for immunosuppression, enabling this therapy to be used in all those with type 1 diabetes.
Collapse
Affiliation(s)
- Sophie Walker
- BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Mahesh Appari
- BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Shareen Forbes
- BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Transplant Unit, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
- Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
7
|
Su W, Feng M, Liu Y, Cao R, Liu Y, Tang J, Pan K, Lan R, Mao Z. ZnT8 Deficiency Protects From APAP-Induced Acute Liver Injury by Reducing Oxidative Stress Through Upregulating Hepatic Zinc and Metallothioneins. Front Pharmacol 2021; 12:721471. [PMID: 34413780 PMCID: PMC8369884 DOI: 10.3389/fphar.2021.721471] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/19/2021] [Indexed: 11/29/2022] Open
Abstract
Zinc transporter 8 (ZnT8) is an important zinc transporter highly expressed in pancreatic islets. Deficiency of ZnT8 leads to a marked decrease in islet zinc, which is thought to prevent liver diseases associated with oxidative stress. Herein, we aimed to investigate whether loss of islet zinc affects the antioxidant capacity of the liver and acute drug-induced liver injury. To address this question, we treated ZnT8 knockout (KO) or wild-type control mice with 300 mg/ kg acetaminophen (APAP) or phosphate-buffered saline (PBS). Unexpectedly, we found that loss of ZnT8 in mice ameliorated APAP-induced injury and was accompanied by inhibition of c-Jun N-terminal kinase (JNK) activation, reduced hepatocyte death, and decreased serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST). An increase in hepatic glutathione (GSH) was observed, corresponding to a decrease in malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE) levels. APAP-induced inflammation and glycogen depletion were alleviated. In contrast, no significant changes were observed in cytochrome P450 family 2 subfamily E member 1 (CYP2E1), the main enzyme responsible for drug metabolism. Elevated levels of hepatic zinc and metallothionein (MT) were also observed, which may contribute to the hepatoprotective effect in ZnT8 KO mice. Taken together, these results suggest that ZnT8 deficiency protects the liver from APAP toxicity by attenuating oxidative stress and promoting hepatocyte proliferation. This study provides new insights into the functions of ZnT8 and zinc as key mediators linking pancreatic and hepatic functions.
Collapse
Affiliation(s)
- Wen Su
- School of Basic Medical Sciences, Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Mingji Feng
- School of Basic Medical Sciences, Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Yuan Liu
- School of Basic Medical Sciences, Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Rong Cao
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yiao Liu
- School of Basic Medical Sciences, Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Junyao Tang
- School of Basic Medical Sciences, Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Ke Pan
- Institute for Advanced Study, Shenzhen University, Shenzhen, China
| | - Rongfeng Lan
- Department of Cell Biology and Medical Genetics, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhuo Mao
- School of Basic Medical Sciences, Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
8
|
Emerging Roles of Metallothioneins in Beta Cell Pathophysiology: Beyond and Above Metal Homeostasis and Antioxidant Response. BIOLOGY 2021; 10:biology10030176. [PMID: 33652748 PMCID: PMC7996892 DOI: 10.3390/biology10030176] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022]
Abstract
Simple Summary Defective insulin secretion by pancreatic beta cells is key for the development of type 2 diabetes but the precise mechanisms involved are poorly understood. Metallothioneins are metal binding proteins whose precise biological roles have not been fully characterized. Available evidence indicated that Metallothioneins are protective cellular effectors involved in heavy metal detoxification, metal ion homeostasis and antioxidant defense. This concept has however been challenged by emerging evidence in different medical research fields revealing novel negative roles of Metallothioneins, including in the context of diabetes. In this review, we gather and analyze the available knowledge regarding the complex roles of Metallothioneins in pancreatic beta cell biology and insulin secretion. We comprehensively analyze the evidence showing positive effects of Metallothioneins on beta cell function and survival as well as the emerging evidence revealing negative effects and discuss the possible underlying mechanisms. We expose in parallel findings from other medical research fields and underscore unsettled questions. Then, we propose some future research directions to improve knowledge in the field. Abstract Metallothioneins (MTs) are low molecular weight, cysteine-rich, metal-binding proteins whose precise biological roles have not been fully characterized. Existing evidence implicated MTs in heavy metal detoxification, metal ion homeostasis and antioxidant defense. MTs were thus categorized as protective effectors that contribute to cellular homeostasis and survival. This view has, however, been challenged by emerging evidence in different medical fields revealing novel pathophysiological roles of MTs, including inflammatory bowel disease, neurodegenerative disorders, carcinogenesis and diabetes. In the present focused review, we discuss the evidence for the role of MTs in pancreatic beta-cell biology and insulin secretion. We highlight the pattern of specific isoforms of MT gene expression in rodents and human beta-cells. We then discuss the mechanisms involved in the regulation of MTs in islets under physiological and pathological conditions, particularly type 2 diabetes, and analyze the evidence revealing adaptive and negative roles of MTs in beta-cells and the potential mechanisms involved. Finally, we underscore the unsettled questions in the field and propose some future research directions.
Collapse
|
9
|
Barman S, Srinivasan K. Diabetes and zinc dyshomeostasis: Can zinc supplementation mitigate diabetic complications? Crit Rev Food Sci Nutr 2020; 62:1046-1061. [PMID: 33938330 DOI: 10.1080/10408398.2020.1833178] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Zinc present in the islet cells of the pancreas is crucial for the synthesis, storage, and secretion of insulin. The excretion of large amounts of zinc from the body is reported in diabetic situations. Zinc depletion and increased oxidative stress have a major impact on the pathogenesis of diabetic complications. It would be most relevant to ascertain if intervention with supplemental zinc compensating for its depletion would beneficially mitigate hyperglycemia and the attendant metabolic abnormalities, and secondary complications in diabetes. An exhaustive literature search on this issue indicates: (1) Concurrent hypozincemia and decreased tissue zinc stores in diabetes as a result of its increased urinary excretion and/or decreased intestinal absorption, (2) Several recent experimental studies have documented that supplemental zinc has a potential hypoglycemic effect in the diabetic situation, and also beneficially modulate the attendant metabolic abnormalities and compromised antioxidant status, and (3) Supplemental zinc also alleviates renal lesions, cataract and the risk of cardiovascular disease accompanying diabetes mellitus, and help restore gastrointestinal health in experimental diabetes. These studies have also attempted to identify the precise mechanisms responsible for zinc-mediated beneficial effects in diabetic situation. The evidence discussed in this review highlights that supplemental zinc may significantly contribute to its clinical application in the management of diabetic hyperglycemia and related metabolic abnormalities, and in the alleviation of secondary complications resulting from diabetic oxidative stress.
Collapse
Affiliation(s)
- Susmita Barman
- Department of Biochemistry, CSIR - Central Food Technological Research Institute, Mysore, India
| | - Krishnapura Srinivasan
- Department of Biochemistry, CSIR - Central Food Technological Research Institute, Mysore, India
| |
Collapse
|
10
|
Bensellam M, Shi YC, Chan JY, Laybutt DR, Chae H, Abou-Samra M, Pappas EG, Thomas HE, Gilon P, Jonas JC. Metallothionein 1 negatively regulates glucose-stimulated insulin secretion and is differentially expressed in conditions of beta cell compensation and failure in mice and humans. Diabetologia 2019; 62:2273-2286. [PMID: 31624901 DOI: 10.1007/s00125-019-05008-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/13/2019] [Indexed: 10/25/2022]
Abstract
AIMS/HYPOTHESIS The mechanisms responsible for beta cell compensation in obesity and for beta cell failure in type 2 diabetes are poorly defined. The mRNA levels of several metallothionein (MT) genes are upregulated in islets from individuals with type 2 diabetes, but their role in beta cells is not clear. Here we examined: (1) the temporal changes of islet Mt1 and Mt2 gene expression in mouse models of beta cell compensation and failure; and (2) the role of Mt1 and Mt2 in beta cell function and glucose homeostasis in mice. METHODS Mt1 and Mt2 expression was assessed in islets from: (1) control lean (chow diet-fed) and diet-induced obese (high-fat diet-fed for 6 weeks) mice; (2) mouse models of diabetes (db/db mice) at 6 weeks old (prediabetes) and 16 weeks old (after diabetes onset) and age-matched db/+ (control) mice; and (3) obese non-diabetic ob/ob mice (16-week-old) and age-matched ob/+ (control) mice. MT1E, MT1X and MT2A expression was assessed in islets from humans with and without type 2 diabetes. Mt1-Mt2 double-knockout (KO) mice, transgenic mice overexpressing Mt1 under the control of its natural promoter (Tg-Mt1) and corresponding control mice were also studied. In MIN6 cells, MT1 and MT2 were inhibited by small interfering RNAs. mRNA levels were assessed by real-time RT-PCR, plasma insulin and islet MT levels by ELISA, glucose tolerance by i.p. glucose tolerance tests and overnight fasting-1 h refeeding tests, insulin tolerance by i.p. insulin tolerance tests, insulin secretion by RIA, cytosolic free Ca2+ concentration with Fura-2 leakage resistant (Fura-2 LR), cytosolic free Zn2+ concentration with Fluozin-3, and NAD(P)H by autofluorescence. RESULTS Mt1 and Mt2 mRNA levels were reduced in islets of murine models of beta cell compensation, whereas they were increased in diabetic db/db mice. In humans, MT1X mRNA levels were significantly upregulated in islets from individuals with type 2 diabetes in comparison with non-diabetic donors, while MT1E and MT2A mRNA levels were unchanged. Ex vivo, islet Mt1 and Mt2 mRNA and MT1 and MT2 protein levels were downregulated after culture with glucose at 10-30 mmol/l vs 2-5 mmol/l, in association with increased insulin secretion. In human islets, mRNA levels of MT1E, MT1X and MT2A were downregulated by stimulation with physiological and supraphysiological levels of glucose. In comparison with wild-type (WT) mice, Mt1-Mt2 double-KO mice displayed improved glucose tolerance in association with increased insulin levels and enhanced insulin release from isolated islets. In contrast, isolated islets from Tg-Mt1 mice displayed impaired glucose-stimulated insulin secretion (GSIS). In both Mt1-Mt2 double-KO and Tg-Mt1 models, the changes in GSIS occurred despite similar islet insulin content, rises in cytosolic free Ca2+ concentration and NAD(P)H levels, or intracellular Zn2+ concentration vs WT mice. In MIN6 cells, knockdown of MT1 but not MT2 potentiated GSIS, suggesting that Mt1 rather than Mt2 affects beta cell function. CONCLUSIONS/INTERPRETATION These findings implicate Mt1 as a negative regulator of insulin secretion. The downregulation of Mt1 is associated with beta cell compensation in obesity, whereas increased Mt1 accompanies beta cell failure and type 2 diabetes.
Collapse
Affiliation(s)
- Mohammed Bensellam
- Pôle d'endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 55 - B1.55.06, B-1200, Brussels, Belgium.
| | - Yan-Chuan Shi
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jeng Yie Chan
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - D Ross Laybutt
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Heeyoung Chae
- Pôle d'endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 55 - B1.55.06, B-1200, Brussels, Belgium
| | - Michel Abou-Samra
- Pôle d'endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 55 - B1.55.06, B-1200, Brussels, Belgium
| | - Evan G Pappas
- St Vincent's Institute, Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, Victoria, Australia
| | - Helen E Thomas
- St Vincent's Institute, Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, Victoria, Australia
| | - Patrick Gilon
- Pôle d'endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 55 - B1.55.06, B-1200, Brussels, Belgium
| | - Jean-Christophe Jonas
- Pôle d'endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 55 - B1.55.06, B-1200, Brussels, Belgium.
| |
Collapse
|
11
|
Zhang M, Chen Y, Yang MJ, Fan XR, Xie H, Zhang L, Nie YS, Yan M. Celastrol attenuates renal injury in diabetic rats via MAPK/NF-κB pathway. Phytother Res 2019; 33:1191-1198. [PMID: 30768745 DOI: 10.1002/ptr.6314] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/17/2022]
Abstract
The purpose of this study was to investigate the renal protective effect of celastrol on diabetic rats. Furthermore, the mechanism of its action was discussed whether it was related to MAPK/NF-κB signaling pathway. There were a total of 36 rats. Six rats were randomly chosen as the control group. The remaining 30 rats were given 1% streptozotocin intraperitoneal injection (50 mg/kg) and were randomly divided into five groups: the model control group, the low-dose celastrol group, the high-dose celastrol group, the Tripterygium wilfordii polyglycosides group, and the MAPK/NF-κB inhibitor group. After 4 weeks of continuous administration, 24-hr urine volume, urinary protein, blood urea nitrogen, and serum creatinine content were observed, and hematoxylin-eosin (HE) staining of the kidney and liver were evaluated. p38MAPK was designated by immunohistochemical method, and NF-κB p65 in renal tissue was detected by western blotting. Our results showed that celastrol could not only reduce contents of creatinine and urea nitrogen in blood but also reduce excretion of urinary protein in diabetic rats, improve renal pathological injury, and down-regulate the expression of p38MAPK and NF-κB p65. In conclusion, celastrol could protect kidney of diabetic rats by regulating the signal pathway of MAPK/NF-κB, inhibiting inflammation and delaying renal injury.
Collapse
Affiliation(s)
- Min Zhang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yan Chen
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mei-Ju Yang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xin-Rong Fan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Hui Xie
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Ling Zhang
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yu-Song Nie
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Miao Yan
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Sargsyan E, Cen J, Roomp K, Schneider R, Bergsten P. Identification of early biological changes in palmitate-treated isolated human islets. BMC Genomics 2018; 19:629. [PMID: 30134843 PMCID: PMC6106933 DOI: 10.1186/s12864-018-5008-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 08/14/2018] [Indexed: 12/13/2022] Open
Abstract
Background Long-term exposure to elevated levels of free fatty acids (FFAs) is deleterious for beta-cell function and may contribute to development of type 2 diabetes mellitus (T2DM). Whereas mechanisms of impaired glucose-stimulated insulin secretion (GSIS) in FFA-treated beta-cells have been intensively studied, biological events preceding the secretory failure, when GSIS is accentuated, are poorly investigated. To identify these early events, we performed genome-wide analysis of gene expression in isolated human islets exposed to fatty acid palmitate for different time periods. Results Palmitate-treated human islets showed decline in beta-cell function starting from day two. Affymetrix Human Transcriptome Array 2.0 identified 903 differentially expressed genes (DEGs). Mapping of the genes onto pathways using KEGG pathway enrichment analysis predicted four islet biology-related pathways enriched prior but not after the decline of islet function and three pathways enriched both prior and after the decline of islet function. DEGs from these pathways were analyzed at the transcript level. The results propose that in palmitate-treated human islets, at early time points, protective events, including up-regulation of metallothioneins, tRNA synthetases and fatty acid-metabolising proteins, dominate over deleterious events, including inhibition of fatty acid detoxification enzymes, which contributes to the enhanced GSIS. After prolonged exposure of islets to palmitate, the protective events are outweighed by the deleterious events, which leads to impaired GSIS. Conclusions The study identifies temporal order between different cellular events, which either promote or protect from beta-cell failure. The sequence of these events should be considered when developing strategies for prevention and treatment of the disease. Electronic supplementary material The online version of this article (10.1186/s12864-018-5008-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ernest Sargsyan
- Department of Medical Cell Biology, Uppsala University, Box 571, 75123, Uppsala, Sweden. .,Molecular Neuroscience Group, Institute of Molecular Biology, National Academy of Sciences, 0014, Yerevan, Armenia.
| | - Jing Cen
- Department of Medical Cell Biology, Uppsala University, Box 571, 75123, Uppsala, Sweden
| | - Kirsten Roomp
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, 7 avenue des Hauts fourneaux, 4362 Esch-Belval, Luxembourg City, Luxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, 7 avenue des Hauts fourneaux, 4362 Esch-Belval, Luxembourg City, Luxembourg
| | - Peter Bergsten
- Department of Medical Cell Biology, Uppsala University, Box 571, 75123, Uppsala, Sweden
| |
Collapse
|
13
|
Choi S, Liu X, Pan Z. Zinc deficiency and cellular oxidative stress: prognostic implications in cardiovascular diseases. Acta Pharmacol Sin 2018; 39:1120-1132. [PMID: 29926844 PMCID: PMC6289396 DOI: 10.1038/aps.2018.25] [Citation(s) in RCA: 246] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/28/2018] [Indexed: 12/11/2022]
Abstract
Zinc is an essential nutrient for human health and has anti-oxidative stress and anti-inflammatory functions. The association between zinc deficiency and the development of cardiovascular diseases (CVDs) has been supported by numerous studies. Supplementing zinc can reduce the risk of atherosclerosis and protect against myocardial infarction and ischemia/reperfusion injury. In this review we summarize the evidence in the literature, to consolidate the current knowledge on the dysregulation of zinc homeostasis in CVDs, and to explore the significant roles of the zinc homeostasis-regulatory proteins in cardiac physiology and pathophysiology. Moreover, this review also deliberates on the potential diagnostic and prognostic implications of zinc/zinc homeostasis-associated molecules (ZIP, ZnT, and MTs) in CVDs.
Collapse
|
14
|
Miki A, Ricordi C, Sakuma Y, Yamamoto T, Misawa R, Mita A, Molano RD, Vaziri ND, Pileggi A, Ichii H. Divergent antioxidant capacity of human islet cell subsets: A potential cause of beta-cell vulnerability in diabetes and islet transplantation. PLoS One 2018; 13:e0196570. [PMID: 29723228 PMCID: PMC5933778 DOI: 10.1371/journal.pone.0196570] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 04/16/2018] [Indexed: 01/09/2023] Open
Abstract
Background Type 1 and Type 2 diabetes mellitus (T1DM and T2DM) are caused by beta(β)-cell loss and functional impairment. Identification of mechanisms of β-cell death and therapeutic interventions to enhance β-cell survival are essential for prevention and treatment of diabetes. Oxidative stress is a common feature of both T1DM and T2DM; elevated biomarkers of oxidative stress are detected in blood, urine and tissues including pancreas of patients with DM. Islet transplantation is a promising treatment for diabetes. However, exposure to stress (chemical and mechanical) and ischemia-reperfusion during isolation and transplantation causes islet loss by generation of reactive oxygen species (ROS). Human intracellular antioxidant enzymes and related molecules are essential defenses against ROS. Antioxidant enzyme levels including superoxide dismutase (SOD), catalase, and glutathione peroxidase (GPX) have been shown to be low in islet cells. However, little is known about the expression and function of antioxidant enzymes within islet cell subsets. We evaluated the expression of the key antioxidant enzymes in β- and alpha(α)-cell and accessed effects of oxidative stress, islet isolation and transplantation on β/α-cell ratio and viability in human islets. Methods Human pancreata from T1DM, T2DM and non-diabetic deceased donors were obtained and analyzed by confocal microscopy. Isolated islets were (I) transplanted in the renal sub-capsular space of streptozotocin-induced diabetic nude mice (in vivo bioassay), or (II) exposed to oxidative (H2O2) and nitrosative (NO donor) stress for 24 hrs in vitro. The ratio, % viability and death of β- and α-cells, and DNA damage (8OHdG) were measured. Results and conclusions Catalase and GPX expression was much lower in β- than α-cells. The β/α-cell ratio fells significantly following islet isolation and transplantation. Exposure to oxidative stress caused a significantly lower survival and viability, with higher DNA damage in β- than α-cells. These findings identified the weakness of β-cell antioxidant capacity as a main cause of vulnerability to oxidative stress. Potential strategies to enhance β-cell antioxidant capacity might be effective in prevention/treatment of diabetes.
Collapse
Affiliation(s)
- Atsushi Miki
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Camillo Ricordi
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Yasunaru Sakuma
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Toshiyuki Yamamoto
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Ryosuke Misawa
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Atsuyoshi Mita
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Ruth D Molano
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Nosratola D Vaziri
- Department of Medicine, University of California, Irvine, United States of America
| | - Antonello Pileggi
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Hirohito Ichii
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America.,Department of Surgery, University of California, Irvine, United States of America
| |
Collapse
|
15
|
Sell DR, Nemet I, Liang Z, Monnier VM. Evidence of glucuronidation of the glycation product LW-1: tentative structure and implications for the long-term complications of diabetes. Glycoconj J 2018; 35:177-190. [PMID: 29305779 DOI: 10.1007/s10719-017-9810-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/01/2017] [Indexed: 01/06/2023]
Abstract
LW-1 is a collagen-linked blue fluorophore whose skin levels increase with age, diabetes and end-stage renal disease (ESRD), and correlate with the long-term progression of microvascular disease and indices of subclinical cardiovascular disease in type 1 diabetes. The chemical structure of LW-1 is still elusive, but earlier NMR analyses showed it has a lysine residue in an aromatic ring coupled to a sugar molecule reminiscent of advanced glycation end-products (AGEs). We hypothesized and demonstrate here that the unknown sugar is a N-linked glucuronic acid. LW-1 was extracted and highly purified from ~99 g insoluble skin collagen obtained at autopsy from patients with diabetes/ESRD using multiple rounds of proteolytic digestion and purification by liquid chromatography (LC). Advanced NMR techniques (1H-NMR, 13C-NMR, 1H-13C HSQC, 1H-1H TOCSY, 1H-13C HMBC) together with LC-mass spectrometry (MS) revealed a loss of 176 amu (atomic mass unit) unequivocally point to the presence of a glucuronic acid moiety in LW-1. To confirm this data, LW-1 was incubated with β-glycosidases (glucosidase, galactosidase, glucuronidase) and products were analyzed by LC-MS. Only glucuronidase could cleave the sugar from the parent molecule. These results establish LW-1 as a glucuronide, now named glucuronidine, and for the first time raise the possible existence of a "glucuronidation pathway of diabetic complications". Future research is needed to rigorously probe this concept and elucidate the molecular origin and biological source of a circulating glucuronidine aglycone.
Collapse
Affiliation(s)
- David R Sell
- Department of Pathology, Case Western Reserve University, Wolstein Research Bldg. 5-301, 2103 Cornell Road, Cleveland, OH, 44106, USA.
| | - Ina Nemet
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA
| | - Zhili Liang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Vincent M Monnier
- Department of Pathology, Case Western Reserve University, Wolstein Research Bldg. 5-301, 2103 Cornell Road, Cleveland, OH, 44106, USA. .,Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
16
|
Programming of Cell Resistance to Genotoxic and Oxidative Stress. Biomedicines 2018; 6:biomedicines6010005. [PMID: 29301323 PMCID: PMC5874662 DOI: 10.3390/biomedicines6010005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 12/23/2017] [Accepted: 12/31/2017] [Indexed: 12/23/2022] Open
Abstract
Different organisms, cell types, and even similar cell lines can dramatically differ in resistance to genotoxic stress. This testifies to the wide opportunities for genetic and epigenetic regulation of stress resistance. These opportunities could be used to increase the effectiveness of cancer therapy, develop new varieties of plants and animals, and search for new pharmacological targets to enhance human radioresistance, which can be used for manned deep space expeditions. Based on the comparison of transcriptomic studies in cancer cells, in this review, we propose that there is a high diversity of genetic mechanisms of development of genotoxic stress resistance. This review focused on possibilities and limitations of the regulation of the resistance of normal cells and whole organisms to genotoxic and oxidative stress by the overexpressing of stress-response genes. Moreover, the existing experimental data on the effect of such overexpression on the resistance of cells and organisms to various genotoxic agents has been analyzed and systematized. We suggest that the recent advances in the development of multiplex and highly customizable gene overexpression technology that utilizes the mutant Cas9 protein and the abundance of available data on gene functions and their signal networks open new opportunities for research in this field.
Collapse
|
17
|
Giacconi R, Cai L, Costarelli L, Cardelli M, Malavolta M, Piacenza F, Provinciali M. Implications of impaired zinc homeostasis in diabetic cardiomyopathy and nephropathy. Biofactors 2017; 43:770-784. [PMID: 28845600 DOI: 10.1002/biof.1386] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/12/2017] [Accepted: 07/27/2017] [Indexed: 12/18/2022]
Abstract
Impaired zinc homeostasis is observed in diabetes mellitus (DM2) and its complications. Zinc has a specific role in pancreatic β-cells via insulin synthesis, storage, and secretion. Intracellular zinc homeostasis is tightly controlled by zinc transporters (ZnT and Zip families) and metallothioneins (MT) which modulate the uptake, storage, and distribution of zinc. Several investigations in animal models demonstrate the protective role of MT in DM2 and its cardiovascular or renal complications, while a copious literature shows that a common polymorphism (R325W) in ZnT8, which affects the protein's zinc transport activity, is associated with increased DM2 risk. Emerging studies highlight a role of other zinc transporters in β-cell function, suggesting that targeting them could make a possible contribution in managing the hyperglycemia in diabetic patients. This article summarizes the current findings concerning the role of zinc homeostasis in DM2 pathogenesis and development of diabetic cardiomyopathy and nephropathy and suggests novel therapeutic targets. © 2017 BioFactors, 43(6):770-784, 2017.
Collapse
Affiliation(s)
- Robertina Giacconi
- Translational Research Center of Nutrition and Ageing, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Lu Cai
- Pediatric Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, Louisville, KY, USA
| | - Laura Costarelli
- Translational Research Center of Nutrition and Ageing, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Maurizio Cardelli
- Advanced Technology Center for Aging Research, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Marco Malavolta
- Translational Research Center of Nutrition and Ageing, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Francesco Piacenza
- Translational Research Center of Nutrition and Ageing, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Mauro Provinciali
- Advanced Technology Center for Aging Research, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| |
Collapse
|
18
|
Kim MK, Shin HM, Jung H, Lee E, Kim TK, Kim TN, Kwon MJ, Lee SH, Rhee BD, Park JH. Comparison of pancreatic beta cells and alpha cells under hyperglycemia: Inverse coupling in pAkt-FoxO1. Diabetes Res Clin Pract 2017; 131:1-11. [PMID: 28666105 DOI: 10.1016/j.diabres.2017.05.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/04/2017] [Accepted: 05/16/2017] [Indexed: 01/09/2023]
Abstract
Type 2 diabetes manifests beta cell deficiencies and alpha cell expansion which is consistent with relative insulin deficiency and glucagon oversecretion. The effects of hyperglycemia on alpha cells are not as understood in comparison to beta cells. Hyperglycemia increases oxidative stress, which induces Akt activation or FoxO activation, depending on cell type. Several studies independently reported that FoxO1 translocations in alpha cells and beta cells were opposite. We compared the responses of pancreatic alpha cells and beta cells against hyperglycemia. Alpha TC-1 cells and Beta TC-6 cells were incubated with control (5mM Glucose) or high glucose (33mM Glucose) with or without PI3K inhibitor or FoxO1 inhibitor. We assessed PI3K, pAkt and phosphorylated FoxO1 (pFoxO1) in both cell lines. Immunostaining of BrdU and FoxO1 was detected by green fluorescence microscopy and confocal microscopy. Hyperglycemia and H2O2 decreased PI3K and pAKT in beta cells, but increased them in alpha cells. FoxO1 localizations and pFoxO1 expressions between alpha cells and beta cells were opposite. Proliferation of beta cells was decreased, but alpha cell proliferation was increased under hyperglycemia. Antioxidant enzymes including superoxide dismutase (SOD) and catalase were increased in beta cells and they were reversed with FoxO1 inhibitor treatment. Increased proliferation in alpha cells under hyperglycemia was attenuated with PI3K inhibitor. In conclusion, hyperglycemia increased alpha cell proliferation and glucagon contents which are opposite to beta cells. These differences may be related to contrasting PI3K/pAkt changes in both cells and subsequent FoxO1 modulation.
Collapse
Affiliation(s)
- Mi-Kyung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea; Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, South Korea.
| | - Hyun Mi Shin
- Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, South Korea
| | - HyeSook Jung
- Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, South Korea
| | - EunJu Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Tae Kyoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Tae Nyun Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Min Jeong Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Soon Hee Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Byoung Doo Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Jeong Hyun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea; Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, South Korea
| |
Collapse
|
19
|
Kim MJ, Lee Y, Jon S, Lee DY. PEGylated bilirubin nanoparticle as an anti-oxidative and anti-inflammatory demulcent in pancreatic islet xenotransplantation. Biomaterials 2017; 133:242-252. [DOI: 10.1016/j.biomaterials.2017.04.029] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/12/2017] [Accepted: 04/13/2017] [Indexed: 01/06/2023]
|
20
|
Lee AS, Lee SH, Lee S, Yang BK. Effects of Streptozotocin, Bisphenol A and Diethylstilbestrol on Production of Reactive Oxygen Species and Lipid Peroxidation in the Boar Sperm. ACTA ACUST UNITED AC 2017. [DOI: 10.15616/bsl.2017.23.2.128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- A-Sung Lee
- College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea
| | - Sang-Hee Lee
- Institute of Animal Resources, Kangwon National University, Chuncheon 24341, Korea
| | - Seunghyung Lee
- College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea
| | - Boo-Keun Yang
- College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
21
|
Aroune D, Libdiri F, Leboucher S, Maouche B, Marco S, El-Aoufi S. Changes in the NFκB and E-cadherin expression are associated to diabetic nephropathy in Psammomys obesus. Saudi J Biol Sci 2017; 24:843-850. [PMID: 28490956 PMCID: PMC5415148 DOI: 10.1016/j.sjbs.2016.05.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 04/29/2016] [Accepted: 05/10/2016] [Indexed: 01/23/2023] Open
Abstract
Diabetes mellitus is a major leading cause of end-stage renal failure, characterized by kidney inflammation and glomerular dysfunction, in worldwide. Kidney inflammation is associated to modifications in the expression levels of pro-inflammatory molecules, such as nuclear factor-κB (NFκB) and adhesion molecules, such as E-cadherin, leading to glomerular dysfunction. However, the relationships between these two processes in human diabetic nephropathy remain an open question. Since Psammomys obesus is an ideal animal model to study diabetes mellitus temporal evolution, we have used this model to study the correlation between kidney structural changes and modification on the expression levels of NFκB and E-cadherin over time. We have demonstrated that, after induction of diabetes metillus with a high energy diet (HED), P. obesus develops the characteristic symptoms of human disease. In detail, at the third month nuclear factor NFκB is expressed in the kidney of diabetic P. obesus and structural renal changes, such as mesangial expansion or interstitial fibrosis, are detectable; at 6 months, thickening of glomerular basement membrane, glomerular sclerosis, and tubular atrophy occurs; at 9 months, symptoms of the final stages of the disease, such as down expression of E-cadherin, happens. As a result of these observations we proposed that NFκB activation and E-cadherin down-expression are interlinked on diabetic kidney disease (DKD).
Collapse
Affiliation(s)
- Djamila Aroune
- Laboratoire de Biologie et de Physiologie des Organismes/MMDED, Faculté des Sciences Biologiques, USTHB, El-Alia, Dar El Beida, Algeria
| | - Farid Libdiri
- Laboratoire de Biologie et de Physiologie des Organismes/MMDED, Faculté des Sciences Biologiques, USTHB, El-Alia, Dar El Beida, Algeria
| | - Sophie Leboucher
- Institut Curie, Centre de Recherche, Plateforme d’Histologie, Orsay F-91405, France
| | - Boubekeur Maouche
- Laboratoire de Biologie et de Physiologie des Organismes/MMDED, Faculté des Sciences Biologiques, USTHB, El-Alia, Dar El Beida, Algeria
| | - Sergio Marco
- Institut Curie, Centre de Recherche, Orsay F-91405, France
- INSERM, U1196, Orsay F-91405, France
- CNRS, UMR9187, Orsay F-91405, France
- Univ. Paris-Sud, Université Paris-Saclay, Saint-Aubin F-91190, France
| | - Salima El-Aoufi
- Laboratoire de Biologie et de Physiologie des Organismes/MMDED, Faculté des Sciences Biologiques, USTHB, El-Alia, Dar El Beida, Algeria
| |
Collapse
|
22
|
Sánchez-Lira NMV, Morales-Miranda A, García de la Mora G, León Contreras JC, González-Sánchez I, Valencia N, Cerbón M, Morimoto S. Orcinol derivative compound with antioxidant properties protects Langerhans islets against streptozotocin damage. J Pharm Pharmacol 2017; 69:305-313. [PMID: 28134974 DOI: 10.1111/jphp.12696] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/29/2016] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To design and synthesize an orcinol derivative compound, 3-formyl-2,4-dihydroxy-5,6-dimethyl sodium benzoate (A4), as an antioxidant molecule and to test its effects on oxidative stress in an in-vitro model of apoptosis of pancreatic rat beta cells induced by streptozotocin (STZ). METHODS Scavenger properties of A4 were assessed using its capacity to capture the DPPH radical in vitro. Antiapoptotic properties of A4 were analysed by electron microscopy and TUNEL assay in rat pancreatic islets in a streptozotocin model. KEY FINDINGS The results show that A4 displays antioxidant activity in in-vitro assays and induced a significant reduction in STZ-induced beta cell apoptosis and low ultrastructural damage to cellular organelles in the rat pancreatic islets as evidenced by electronic microscopy, this effect could be attributed to its antioxidant activity in a similar manner than resveratrol. CONCLUSION The overall results indicate that the new orcinol derivative molecule displays both antioxidant and antiapoptotic effects and protect pancreatic beta cells against STZ damage.
Collapse
Affiliation(s)
- Nancy M V Sánchez-Lira
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Angélica Morales-Miranda
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Gustavo García de la Mora
- División de Posgrado, Departamento de Química Orgánica, Facultad de Química, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, Mexico
| | - Juan Carlos León Contreras
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Ignacio González-Sánchez
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, Mexico
| | - Norma Valencia
- Departamento de Farmacia, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Facultad de Química, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Sumiko Morimoto
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| |
Collapse
|
23
|
KOBAYASHI M, KURATA T, HAMANA Y, HIRAMITSU M, INOUE T, MURAI A, HORIO F. Coffee Ingestion Suppresses Hyperglycemia in Streptozotocin-Induced Diabetic Mice. J Nutr Sci Vitaminol (Tokyo) 2017; 63:200-207. [DOI: 10.3177/jnsv.63.200] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Misato KOBAYASHI
- Department of Applied Molecular Bioscience, Graduate School of Bioagricultural Sciences, Nagoya University
| | - Takao KURATA
- Department of Applied Molecular Bioscience, Graduate School of Bioagricultural Sciences, Nagoya University
| | - Yoshiki HAMANA
- Department of Applied Molecular Bioscience, Graduate School of Bioagricultural Sciences, Nagoya University
| | | | - Takashi INOUE
- Research & Development Division, Pokka Sapporo Food & Beverage Ltd
| | - Atsushi MURAI
- Department of Applied Molecular Bioscience, Graduate School of Bioagricultural Sciences, Nagoya University
| | - Fumihiko HORIO
- Department of Applied Molecular Bioscience, Graduate School of Bioagricultural Sciences, Nagoya University
| |
Collapse
|
24
|
Schott-Ohly P, Lgssiar A, Partke HJ, Hassan M, Friesen N, Gleichmann H. Prevention of Spontaneous and Experimentally Induced Diabetes in Mice with Zinc Sulfate-Enriched Drinking Water is Associated with Activation and Reduction of NF-κB and AP-1 in Islets, Respectively. Exp Biol Med (Maywood) 2016; 229:1177-85. [PMID: 15564445 DOI: 10.1177/153537020422901113] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Recently, we reported that zinc sulfate-enriched (25 mM) drinking water (Zn2+) protected male C57BL/6 mice from diabetes induced by multiple low doses of streptozotocin (MLD-STZ) and that MLD-STZ activates the transcription factors nuclear factor (NF)-κB and activator protein (AP)-1 in islets of these mice. Therefore, we studied the effect of Zn2+ on spontaneous diabetes in female nonobese diabetic (NOD) mice and on the activity of NF-κB and AP-1 in islets of NOD and MLD-STZ–injected male C57BL/6 mice. We hypothesized that Zn2+ may affect NF-κB, which may play a key role in immune-mediated diabetogenesis. Here we continuously administered Zn2+ to NOD mice, to both parents and their F1 offspring, and treated C57BL/6 male mice with MLD-STZ either alone or in addition to Zn2+. We assessed effects of Zn2+ on insulitis and peri-insulitis in 8-week-old NOD mice and analyzed NF-κB and AP-1 activities in islets. Zn2+ significantly prevented diabetes in female F1 offspring and significantly reduced insulitis and peri-insulitis. Zn2+ significantly stimulated NF-κB and AP-1 activation in NOD mice, in contrast, in C57BL/6 mice, Zn2+ significantly reduced their activation by MLD-STZ. These data demonstrate that NF-κB may play a critical role in immune-mediated diabetes. Depending on the mode of β-cell destruction, Zn2+ may prevent apoptosis through activation of NF-κB in NOD mice or prevent inflammatory immune destruction through inhibition of NF-κB in MLD-STZ-treated C57BL/6 mice.
Collapse
Affiliation(s)
- Patricia Schott-Ohly
- German Diabetes Center, German Diabetes Research Institute, D-40225 Düsseldorf, Germany
| | | | | | | | | | | |
Collapse
|
25
|
Kim MJ, Hwang YH, Kim YH, Lee DY. Immunomodulation of cell-penetrating tat-metallothionein for successful outcome of xenotransplanted pancreatic islet. J Drug Target 2016; 25:350-359. [PMID: 27829285 DOI: 10.1080/1061186x.2016.1258704] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Pancreatic islet transplantation is a promising treatment for treatment of type 1 diabetes; however, transplantation outcomes have been disappointing due to early graft loss that is mediated by many immune responses. Immune cells not only directly damaged islet but also produced reactive oxygen species (ROS), which is highly toxic to islet cells. Metallothionein (MT) can provide protection against oxidative stress by scavenging various ranges of ROS including superoxide, hydroxyl radical, hydrogen peroxide and nitric oxide. For scavenging immune response-induced ROS, cell-penetrating Tat peptide-metallothionein (Tat-MT) was delivered into islets. The viability of Tat-MT-treated islets was not damaged during co-culture with macrophages or ROS-generating paraquat. When Tat-MT-treated islets were xenotransplanted, ROS production was significantly attenuated at the islets. Eventually, the survival time of Tat-MT-treated islets was significantly enhanced without any immunosuppressant medicine. Additionally, we confirmed that the survival time of Tat-MT-treated islets in all animals was dramatically improved when accompanied with low dose immunosuppressive agents (tacrolimus and anti-CD154 monoclonal antibody), indicating that Tat-MT delivery could have synergistic effect with immunosuppressants. Collectively, this new combination therapy of Tat-MT delivery with low dose immunosuppressant would be a powerful remedy for successful outcome of islet xenotransplantation.
Collapse
Affiliation(s)
- Min Jun Kim
- a Departments of Bioengineering , College of Engineering, and BK21 PLUS Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University , Seoul , Republic of Korea
| | - Yong Hwa Hwang
- a Departments of Bioengineering , College of Engineering, and BK21 PLUS Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University , Seoul , Republic of Korea
| | - Yong Hee Kim
- a Departments of Bioengineering , College of Engineering, and BK21 PLUS Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University , Seoul , Republic of Korea
| | - Dong Yun Lee
- a Departments of Bioengineering , College of Engineering, and BK21 PLUS Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University , Seoul , Republic of Korea.,b Institute of Nano Science and Technology (INST), Hanyang University , Seoul , Republic of Korea
| |
Collapse
|
26
|
Abstract
Zinc (Zn) is important in a number of processes related to insulin secretion and insulin activity in peripheral tissues, making this element an interesting potential co-adjuvant in the treatment of patients with type 2 diabetes (T2D). This issue has been matter of interest in recent years. The available evidence is analyzed in this review. Information from epidemiologic studies evaluating the relationship between Zn and T2D is inconsistent. Furthermore, few studies examined the association between Zn status and insulin action and/or glucose homeostasis. In terms of usefulness of Zn as a preventive agent for T2D development, information is insufficient to reach firm conclusions. Results from Zn supplementation trials found some positive effects only in those with initial sub normal Zn status in a significant proportion of individuals. In conclusion, the effect of Zn on patients with type 2 diabetes is still an open question, and better study designs are needed to clarify the real impact and characteristics of the Zn-diabetes interaction.
Collapse
Affiliation(s)
- Manuel Ruz
- Department of Nutrition, Faculty of Medicine, University of Chile, Independencia 1027, Postal Code 8380453, Santiago, Chile.
| | - Fernando Carrasco
- Department of Nutrition, Faculty of Medicine, University of Chile, Independencia 1027, Postal Code 8380453, Santiago, Chile
| | - Andrés Sánchez
- Department of Nutrition, Faculty of Medicine, University of Chile, Independencia 1027, Postal Code 8380453, Santiago, Chile
| | - Alvaro Perez
- Department of Nutrition, Faculty of Medicine, University of Chile, Independencia 1027, Postal Code 8380453, Santiago, Chile
| | - Pamela Rojas
- Department of Nutrition, Faculty of Medicine, University of Chile, Independencia 1027, Postal Code 8380453, Santiago, Chile
| |
Collapse
|
27
|
A Single-Cell Transcriptome Atlas of the Human Pancreas. Cell Syst 2016; 3:385-394.e3. [PMID: 27693023 PMCID: PMC5092539 DOI: 10.1016/j.cels.2016.09.002] [Citation(s) in RCA: 798] [Impact Index Per Article: 88.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 07/04/2016] [Accepted: 09/07/2016] [Indexed: 11/21/2022]
Abstract
To understand organ function, it is important to have an inventory of its cell types and of their corresponding marker genes. This is a particularly challenging task for human tissues like the pancreas, because reliable markers are limited. Hence, transcriptome-wide studies are typically done on pooled islets of Langerhans, obscuring contributions from rare cell types and of potential subpopulations. To overcome this challenge, we developed an automated platform that uses FACS, robotics, and the CEL-Seq2 protocol to obtain the transcriptomes of thousands of single pancreatic cells from deceased organ donors, allowing in silico purification of all main pancreatic cell types. We identify cell type-specific transcription factors and a subpopulation of REG3A-positive acinar cells. We also show that CD24 and TM4SF4 expression can be used to sort live alpha and beta cells with high purity. This resource will be useful for developing a deeper understanding of pancreatic biology and pathophysiology of diabetes mellitus. Single-cell sequencing of human pancreas allows in silico purification of cell types We provide cell-type-specific genes, transcription factors, and cell-surface markers StemID finds outlier populations of acinar and beta cells CD24 and TM4SF4 function as two markers to enrich for alpha and beta cells
Collapse
|
28
|
Salemi Z, Rafie E, Goodarzi MT, Ghaffari MA. Effect of Metformin, Acarbose and Their Combination on the Serum Visfatin Level in Nicotinamide/Streptozocin-Induced Type 2 Diabetic Rats. IRANIAN RED CRESCENT MEDICAL JOURNAL 2016; 18:e23814. [PMID: 27247792 PMCID: PMC4884299 DOI: 10.5812/ircmj.23814] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/15/2014] [Accepted: 11/11/2014] [Indexed: 01/06/2023]
Abstract
BACKGROUND Diabetes mellitus is a chronic metabolic disease with life-threatening complications. Metformin and acarbose are two oral antidiabetic drugs. OBJECTIVES This experimental study was designed and carried out at the Arak University of Medical Sciences in Arak, Iran, to investigate the effects of these drugs (both alone and in combination) on glycemic control, lipid profile, and serum visfatin levels in nicotinamide/streptozotocin type 2 diabetic rats. MATERIALS AND METHODS Type 2 diabetes was induced in 30 male Wistar rats by the administration of streptozotocin (STZ) (60 mg/kg body weight) intraperitoneally (IP) 15 minutes after the IP administration of nicotinamide (110 mg/kg body weight). After one week, the diabetic rats were randomly divided into four groups. Three diabetic groups were treated with 150 mg/kg/day of metformin, acarbose (40 mg/100 g of diet), or a combination of the two for six weeks, respectively. Biochemical parameters, including fasting blood glucose, glycated hemoglobin, lipid profile, insulin, and visfatin were assessed and compared with those of the control diabetic group. RESULTS The data showed metformin, acarbose, and acarbose + metformin downregulated visfatin levels in diabetic rats, but only the reduction in metformin-treated rats was significant (162 ± 21.7, 195.66 ± 6.45 (ng/l), P = 0.001). Fasting blood glucose and glycated hemoglobin decreased significantly in all treated rats, specifically in the treated group that received the two drugs in combination. The serum insulin level was also reduced in all treated groups, and it was significant in the acarbose (P < 0.05) and the combination therapy groups (P < 0.05). The lipid profile improved in all treated groups. CONCLUSIONS Compared with acarbose or metformin monotherapy, the addition of acarbose to metformin had superior antihyperglycemia efficacy and provided an efficacious and safe alternative for the treatment of type 2 diabetic rats. Acarbose/metformin reduced the fasting blood glucose and glycated hemoglobin without significant changes in serum visfatin levels.
Collapse
Affiliation(s)
- Zahra Salemi
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, IR Iran
- Department of Biochemistry, Arak University of Medical Sciences, Arak, IR Iran
- Corresponding Author: Zahra Salemi, Department of Biochemistry, Arak University of Medical Sciences, Arak, IR Iran. Tel: +98-9183645842, Fax: +98-8634173529, E-mail:
| | - Elham Rafie
- Department of Biochemistry, Arak University of Medical Sciences, Arak, IR Iran
| | - Mohamad Taghi Goodarzi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, IR Iran
| | - Mohamad ali Ghaffari
- Department of Biochemistry, Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| |
Collapse
|
29
|
Dziegiel P, Pula B, Kobierzycki C, Stasiolek M, Podhorska-Okolow M. Metallothioneins in Normal and Cancer Cells. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2016; 218:1-117. [PMID: 26847563 DOI: 10.1007/978-3-319-27472-0_1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Metallothioneins (MTs) are low molecular weight proteins, which are present in almost all types of organisms. In mammals, four main MT isoforms designated from MT-1 to MT-4 were identified. Their biological role, according to their characteristic structure, was shown to be mostly associated with cellular metabolism of metal ions, especially zinc. Moreover, the available evidence suggests broad regulatory properties of MTs in the control of cell senescence and various pathological processes including neurodegeneration, cardiovascular pathology, metabolic disorders, and various malignancies. This extensive review provides general in formation on the structure of MT family members and the cellular functions of MT-1, MT-2, and MT-4 isoforms as well as insights into divergent biological roles of MT-3. Due to the involvement of MT molecules in various processes related to carcinogenesis, an organ-specific presentation of current data concerning their potential impact on the progression of various tumors is given. The regulatory role of MT family members in the function of the immune system is also discussed in depth.
Collapse
Affiliation(s)
- Piotr Dziegiel
- Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland.,Department of Physiotherapy, Wroclaw University School of Physical Education, Wroclaw, Poland
| | - Bartosz Pula
- Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland
| | - Christopher Kobierzycki
- Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland.,Department of Physiotherapy, Wroclaw University School of Physical Education, Wroclaw, Poland
| | - Mariusz Stasiolek
- Department of Neurology, Polish Mother's Memorial Hospital-Research Institute, Lodz, Poland
| | | |
Collapse
|
30
|
Bruin JE, Asadi A, Fox JK, Erener S, Rezania A, Kieffer TJ. Accelerated Maturation of Human Stem Cell-Derived Pancreatic Progenitor Cells into Insulin-Secreting Cells in Immunodeficient Rats Relative to Mice. Stem Cell Reports 2015; 5:1081-1096. [PMID: 26677767 PMCID: PMC4682152 DOI: 10.1016/j.stemcr.2015.10.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/22/2015] [Accepted: 10/23/2015] [Indexed: 11/30/2022] Open
Abstract
Pluripotent human embryonic stem cells (hESCs) are a potential source of transplantable cells for treating patients with diabetes. To investigate the impact of the host recipient on hESC-derived pancreatic progenitor cell maturation, cells were transplanted into immunodeficient SCID-beige mice or nude rats. Following the transplant, basal human C-peptide levels were consistently higher in mice compared with rats, but only rats showed robust meal- and glucose-responsive human C-peptide secretion by 19-21 weeks. Grafts from rats contained a higher proportion of insulin:glucagon immunoreactivity, fewer exocrine cells, and improved expression of mature β cell markers compared with mice. Moreover, ECM-related genes were enriched, the collagen network was denser, and blood vessels were more intricately integrated into the engrafted endocrine tissue in rats relative to mice. Overall, hESC-derived pancreatic progenitor cells matured faster in nude rats compared with SCID-beige mice, indicating that the host recipient can greatly influence the fate of immature pancreatic progenitor cells post-transplantation.
Collapse
Affiliation(s)
- Jennifer E Bruin
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Ali Asadi
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Jessica K Fox
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Suheda Erener
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Alireza Rezania
- BetaLogics Venture, Janssen R&D LLC, 1000 Route 202 South, Room J108A, Raritan, NJ 08869, USA
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Surgery, University of British Columbia, 950 West 10(th) Avenue, Vancouver, BC V5Z 1M9, Canada.
| |
Collapse
|
31
|
Challenges and issues with streptozotocin-induced diabetes - A clinically relevant animal model to understand the diabetes pathogenesis and evaluate therapeutics. Chem Biol Interact 2015; 244:49-63. [PMID: 26656244 DOI: 10.1016/j.cbi.2015.11.032] [Citation(s) in RCA: 254] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 11/18/2015] [Accepted: 11/26/2015] [Indexed: 02/07/2023]
Abstract
Streptozotocin (STZ) has been extensively used over the last three decades to induce diabetes in various animal species and to help screen for hypoglycemic drugs. STZ induces clinical features in animals that resemble those associated with diabetes in humans. For this reason STZ treated animals have been used to study diabetogenic mechanisms and for preclinical evaluation of novel antidiabetic therapies. However, the physiochemical characteristics and associated toxicities of STZ are still major obstacles for researchers using STZ treated animals to investigate diabetes. Another major challenges in STZ-induced diabetes are sustaining uniformity, suitability, reproducibility and induction of diabetes with minimal animal lethality. Lack of appropriate use of STZ was found to be associated with increased mortality and animal suffering. During STZ use in animals, attention should be paid to several factors such as method of preparation of STZ, stability, suitable dose, route of administration, diet regimen, animal species with respect to age, body weight, gender and the target blood glucose level used to represent hyperglycemia. Therefore, protocol for STZ-induced diabetes in experimental animals must be meticulously planned. This review highlights specific skills and strategies involved in the execution of STZ-induced diabetes model. The present review aims to provide insight into diabetogenic mechanisms of STZ, specific toxicity of STZ with its significance and factors responsible for variations in diabetogenic effects of STZ. Further this review also addresses ways to minimize STZ-induced mortality, suggests methods to improve STZ-based experimental models and best utilize them for experimental studies purported to understand diabetes pathogenesis and preclinical evaluation of drugs.
Collapse
|
32
|
Chen S, Han J, Liu Y. Dual Opposing Roles of Metallothionein Overexpression in C57BL/6J Mouse Pancreatic β-Cells. PLoS One 2015; 10:e0137583. [PMID: 26335571 PMCID: PMC4559390 DOI: 10.1371/journal.pone.0137583] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 08/18/2015] [Indexed: 11/20/2022] Open
Abstract
Background Growing evidence indicates that oxidative stress (OS), a persistent state of excess amounts of reactive oxygen species (ROS) along with reactive nitrogen species (RNS), plays an important role in insulin resistance, diabetic complications, and dysfunction of pancreatic β-cells. Pancreatic β-cells contain exceptionally low levels of antioxidant enzymes, rendering them susceptible to ROS-induced damage. Induction of antioxidants has been proposed to be a way for protecting β-cells against oxidative stress. Compared to other antioxidants that act against particular β-cell damages, metallothionein (MT) is the most effective in protecting β-cells from several oxidative stressors including nitric oxide, peroxynitrite, hydrogen peroxide, superoxide and streptozotocin (STZ). We hypothesized that MT overexpression in pancreatic β-cells would preserve β-cell function in C57BL/6J mice, an animal model susceptible to high fat diet-induced obesity and type 2 diabetes. Research Design and Methods The pancreatic β-cell specific MT overexpression was transferred to C57BL/6J background by backcrossing. We studied transgenic MT (MT-tg) mice and wild-type (WT) littermates at 8 weeks and 18 weeks of age. Several tests were performed to evaluate the function of islets, including STZ in vivo treatment, intraperitoneal glucose tolerance tests (IPGTT) and plasma insulin levels during IPGTT, pancreatic and islet insulin content measurement, insulin secretion, and islet morphology assessment. Gene expression in islets was performed by quantitative real-time PCR and PCR array analysis. Protein levels in pancreatic sections were evaluated by using immunohistochemistry. Results The transgenic MT protein was highly expressed in pancreatic islets. MT-tg overexpression significantly protected mice from acute STZ-induced ROS at 8 weeks of age; unexpectedly, however, MT-tg impaired glucose stimulated insulin secretion (GSIS) and promoted the development of diabetes. Pancreatic β-cell function was significantly impaired, and islet morphology was also abnormal in MT-tg mice, and more severe damage was detected in males. The unique gene expression pattern and abnormal protein levels were observed in MT-tg islets. Conclusions MT overexpression protected β-cells from acute STZ-induced ROS damages at young age, whereas it impaired GSIS and promoted the development of diabetes in adult C57BL/6J mice, and more severe damage was found in males.
Collapse
Affiliation(s)
- Suqin Chen
- The Research Institute for Children, Children’s Hospital at New Orleans, New Orleans, Louisiana, United States of America
- Department of Medical Genetics, Zhongshan Medical College, Sun Yat-sen University, Guangzhou, Guangdong Province, People’s Republic of China
- * E-mail:
| | - Junying Han
- The Research Institute for Children, Children’s Hospital at New Orleans, New Orleans, Louisiana, United States of America
| | - Yeqi Liu
- The Research Institute for Children, Children’s Hospital at New Orleans, New Orleans, Louisiana, United States of America
- Department of Pediatrics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| |
Collapse
|
33
|
McCarty MF, DiNicolantonio JJ. The protection conferred by chelation therapy in post-MI diabetics might be replicated by high-dose zinc supplementation. Med Hypotheses 2015; 84:451-5. [DOI: 10.1016/j.mehy.2015.01.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 01/23/2015] [Accepted: 01/26/2015] [Indexed: 12/20/2022]
|
34
|
Byun HR, Choi JA, Koh JY. The role of metallothionein-3 in streptozotocin-induced beta-islet cell death and diabetes in mice. Metallomics 2015; 6:1748-57. [PMID: 25054451 DOI: 10.1039/c4mt00143e] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Metallothionein-3 (Mt3), a zinc (Zn)-regulatory protein mainly expressed in the central nervous system, may contribute to oxidative cell death. In the present study, we examined the possible role of Mt3 in streptozotocin (STZ)-induced islet cell death and consequent hyperglycemia. Quantitative real-time polymerase chain reaction (RT-PCR) confirmed that islet cells expressed Mt3 mRNA. In all cases, wild-type (WT) mice injected with STZ exhibited hyperglycemia 7-21 days later. In stark contrast, all Mt3-null mice remained normoglycemic following STZ injection. STZ treatment increased free Zn levels in islet cells and induced their death in WT mice, but failed to do so in Mt3-null mice. Consistent with this, cultured Mt3-null islet cells exhibited striking resistance to STZ toxicity. Notably, PDE3a (phosphodiesterase 3A) was downregulated in islets of Mt3-null mice compared to those of WT mice, and was not induced by STZ treatment. Moreover, the PDE3 inhibitor cilostazol reduced islet cell death, likely by increasing cAMP levels, further supporting a role for PDE3 in STZ-induced islet cell death. Collectively, these results demonstrate that Mt3 may act through PDE3a to play a key role in Zn dyshomeostasis and cell death in STZ-treated islets.
Collapse
Affiliation(s)
- Hyae-Ran Byun
- Neural Injury Research Center, Asan Institute for Life Science, University of Ulsan College of Medicine, Seoul, Korea
| | | | | |
Collapse
|
35
|
Nygaard SB, Lund NS, Larsen A, Pedersen N, Rungby J, Smidt K. Exogenous metallothionein potentiates the insulin response at normal glucose concentrations in INS-1E beta-cells without disturbing intracellular ZnT8 expression. Basic Clin Pharmacol Toxicol 2014; 116:173-7. [PMID: 24964825 DOI: 10.1111/bcpt.12287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/10/2014] [Indexed: 12/27/2022]
Affiliation(s)
- Sanne B Nygaard
- Department of Biomedicine - Pharmacology, Aarhus University, Aarhus, Denmark
| | | | | | | | | | | |
Collapse
|
36
|
Kaur K, Gupta R, Saraf SA, Saraf SK. Zinc: The Metal of Life. Compr Rev Food Sci Food Saf 2014; 13:358-376. [PMID: 33412710 DOI: 10.1111/1541-4337.12067] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 02/05/2014] [Indexed: 01/15/2023]
Abstract
The importance of zinc was 1st reported for Aspergillus niger. It took over 75 y to realize that zinc is also an essential trace element for rats, and an additional 30 y went by before it was recognized that this was also true for humans. The adult body contains about 2 to 3 g of zinc. Zinc is found in organs, tissues, bones, fluids, and cells. It is essential for many physiological functions and plays a significant role in a number of enzyme actions in the living systems. Bioinformatics estimates report that 10% of the human proteome contains zinc-binding sites. Based on its role in such a plethora of cellular components, zinc has diverse biological functions from enzymatic catalysis to playing a crucial role in cellular neuronal systems. Thus, based on the various published studies and reports, it is pertinent to state that zinc is one of the most important essential trace metals in human nutrition and lifestyle. Its deficiency may severely affect the homeostasis of a biological system. This review compiles the role of zinc in prophylaxis/therapeutics and provides current information about its effect on living beings.
Collapse
Affiliation(s)
- Kuljeet Kaur
- Faculty of Pharmacy, Babu Banarasi Das Natl. Inst. of Technology and Management (BBD Univ.), Lucknow, India
| | - Rajiv Gupta
- Faculty of Pharmacy, Babu Banarasi Das Natl. Inst. of Technology and Management (BBD Univ.), Lucknow, India
| | - Shubhini A Saraf
- Dept. of Pharmaceutical Sciences, SB&BT, Babasaheb Bhimrao Ambedkar Univ., Lucknow, India
| | - Shailendra K Saraf
- Faculty of Pharmacy, Babu Banarasi Das Northern India Inst. of Technology, Lucknow, India
| |
Collapse
|
37
|
Zhang X, Zhao Y, Chu Q, Wang ZY, Li H, Chi ZH. Zinc modulates high glucose-induced apoptosis by suppressing oxidative stress in renal tubular epithelial cells. Biol Trace Elem Res 2014; 158:259-67. [PMID: 24591003 DOI: 10.1007/s12011-014-9922-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 01/28/2014] [Indexed: 11/25/2022]
Abstract
Hyperglycemia is a characteristic of diabetic nephropathy, inducing renal tubular cell apoptosis by eliciting oxidative stress and inflammation. Zinc (Zn) is known as an essential trace element in many enzymes and proteins involved in antioxidant defenses, electron transport, and exerting antiapoptotic or cytoprotective effects. In this study, the underlying mechanisms involved in the protective effects of Zn on high glucose-induced cytotoxicity were explored using cultured renal tubular epithelial cells (NRK-52E). The authors discovered that Zn supplementation inhibited high glucose (HG)-induced NRK-52E cell apoptosis by attenuating reactive oxygen species production, inhibiting HG-induced caspase-3 and caspase-9 activation, and inhibiting the release of cytochrome c from mitochondria to the cytosol. Further analysis revealed that Zn supplementation facilitated cell survival through increasing nuclear translocation of NF-E2-related factor 2 (Nrf2), leading to increased regulation of levels of two antioxidant enzymes, hemeoxygenase-1 and glutamate cysteine ligase, which provided an adaptive survival response against the HG-induced oxidative cytotoxicity. Moreover, the Zn-mediated increases in Nrf2 activity were suppressed by the pharmacological inhibition of Akt or extracellular signal-regulated kinase 1/2. Taken together, these findings suggest that Zn antiapoptosis capacity through the activation of Akt and ERK signal pathways leads to Nrf2 activation and, subsequently, Nrf2 target gene induction, thereby protecting the NRK-52E cells from HG-induced apoptosis.
Collapse
Affiliation(s)
- Xiuli Zhang
- Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, Liaoning, People's Republic of China
| | | | | | | | | | | |
Collapse
|
38
|
Nygaard SB, Larsen A, Knuhtsen A, Rungby J, Smidt K. Effects of zinc supplementation and zinc chelation on in vitro β-cell function in INS-1E cells. BMC Res Notes 2014; 7:84. [PMID: 24502363 PMCID: PMC3923740 DOI: 10.1186/1756-0500-7-84] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 02/04/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Zinc is essential for the activities of pancreatic β-cells, especially insulin storage and secretion. Insulin secretion leads to co-release of zinc which contributes to the paracrine communication in the pancreatic islets. Zinc-transporting proteins (zinc-regulated transporter, iron-regulated transporter-like proteins [ZIPs] and zinc transporters [ZnTs]) and metal-buffering proteins (metallothioneins, MTs) tightly regulate intracellular zinc homeostasis. The present study investigated how modulation of cellular zinc availability affects β-cell function using INS-1E cells. RESULTS Using INS-1E cells, we found that zinc supplementation and zinc chelation had significant effects on insulin content and insulin secretion. Supplemental zinc within the physiological concentration range induced insulin secretion. Insulin content was reduced by zinc chelation with N,N,N',N-tektrakis(2-pyridylmethyl)-ethylenediamine. The changes in intracellular insulin content following exposure to various concentrations of zinc were reflected by changes in the expression patterns of MT-1A, ZnT-8, ZnT-5, and ZnT-3. Furthermore, high zinc concentrations induced cell necrosis while zinc chelation induced apoptosis. Finally, cell proliferation was sensitive to changes in zinc the concentration. CONCLUSION These results indicate that the β-cell-like function and survival of INS-1E cells are dependent on the surrounding zinc concentrations. Our results suggest that regulation of zinc homeostasis could represent a pharmacological target.
Collapse
Affiliation(s)
- Sanne Bjørn Nygaard
- Department of Biomedicine, Centre of Pharmacology and Pharmacotherapy, Health, Aarhus University, Wilhelm Meyers Allé 4, Bld 1240, Aarhus, 8000, Denmark.
| | | | | | | | | |
Collapse
|
39
|
Hypoxic resistance of hypodermically transplanted pancreatic islets by using cell-absorbable antioxidant Tat-metallothionein. J Control Release 2013; 172:1092-101. [DOI: 10.1016/j.jconrel.2013.09.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 09/13/2013] [Accepted: 09/26/2013] [Indexed: 11/22/2022]
|
40
|
Lee J, Sugiyama T, Liu Y, Wang J, Gu X, Lei J, Markmann JF, Miyazaki S, Miyazaki JI, Szot GL, Bottino R, Kim SK. Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. eLife 2013; 2:e00940. [PMID: 24252877 PMCID: PMC3826580 DOI: 10.7554/elife.00940] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Pancreatic islet β-cell insufficiency underlies pathogenesis of diabetes mellitus; thus, functional β-cell replacement from renewable sources is the focus of intensive worldwide effort. However, in vitro production of progeny that secrete insulin in response to physiological cues from primary human cells has proven elusive. Here we describe fractionation, expansion and conversion of primary adult human pancreatic ductal cells into progeny resembling native β-cells. FACS-sorted adult human ductal cells clonally expanded as spheres in culture, while retaining ductal characteristics. Expression of the cardinal islet developmental regulators Neurog3, MafA, Pdx1 and Pax6 converted exocrine duct cells into endocrine progeny with hallmark β-cell properties, including the ability to synthesize, process and store insulin, and secrete it in response to glucose or other depolarizing stimuli. These studies provide evidence that genetic reprogramming of expandable human pancreatic cells with defined factors may serve as a general strategy for islet replacement in diabetes. DOI:http://dx.doi.org/10.7554/eLife.00940.001 Diabetes mellitus is a disease that can lead to dangerously high blood sugar levels, causing numerous complications such as heart disease, glaucoma, skin disorders, kidney disease, and nerve damage. In healthy individuals, beta cells in the pancreas produce a hormone called insulin, which stimulates cells in the liver, muscles and fat to take up glucose from the blood. However, this process is disrupted in people with diabetes, who either have too few pancreatic beta cells (type 1 diabetes) or do not respond appropriately to insulin (type 2 diabetes). All patients with type 1 diabetes, and some with type 2, must inject themselves regularly with insulin, but this does not always fully control the disease. Some type 1 patients have been successfully treated with beta cells transplanted from deceased donors, but there are not enough donor organs available for this to become routine. Thus, intensive efforts worldwide are focused on generating insulin-producing cells in the lab from human stem cells. However, the cells produced in this way can give rise to tumors. Now, Lee et al. have shown that duct cells, which make up about 30% of the human pancreas, can be converted into cells capable of producing and secreting insulin. Ductal cells obtained from donor pancreases were first separated from the remaining tissue and grown in cell culture. Viruses were then used to introduce genes that reprogrammed the ductal cells so that they acquired the ability to make, process and store insulin, and to release it in response to glucose—hallmark features of functional beta cells. As well as providing a potential source of cells for use in transplant or cell conversion therapies for diabetes, the ability to grow and maintain human pancreatic ductal cells in culture may make it easier to study other diseases that affect the pancreas, including pancreatitis, cystic fibrosis, and adenocarcinoma. DOI:http://dx.doi.org/10.7554/eLife.00940.002
Collapse
Affiliation(s)
- Jonghyeob Lee
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, United States
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ruz M, Carrasco F, Rojas P, Codoceo J, Inostroza J, Basfi-fer K, Valencia A, Vásquez K, Galgani J, Pérez A, López G, Arredondo M, Perez-Bravo F. Zinc as a potential coadjuvant in therapy for type 2 diabetes. Food Nutr Bull 2013; 34:215-21. [PMID: 23964394 DOI: 10.1177/156482651303400210] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Type 2 diabetes is highly prevalent in populations having high rates of overweight and obesity. It is a chronic condition responsible for long-term severe dysfunction of several organs, including the kidneys, heart, blood vessels, and eyes. Although there are a number of pharmacologic products in the market to treat insulin resistance and impaired insulin secretion--the most prominent features of this disease--interventions directed at preserving the integrity and function of beta-cells in the long term are less available. The use of some nutrients with important cellular protective roles that may lead to a preservation of beta-cells has not been fully tested; among these, zinc may be an interesting candidate. OBJECTIVE To assess the potential of zinc supplementation as coadjuvant to diabetes therapy. METHODS This article reviews the available information on the use of zinc as part of diabetes therapy. RESULTS Cellular and animal models provide information on the insulin mimetic action of zinc, as well as its role as a regulator of oxidative stress, inflammation, apoptosis, and insulin secretion. Zinc supplementation studies in humans are limited, although some positive effects have been reported; mainly, a modest but significant reduction in fasting glucose and a trend to decreased glycated hemoglobin (HbA1c). CONCLUSIONS Zinc supplementation may have beneficial effects on glycemic control. Nevertheless, among the studies considered, the vast majority lasted for 6 months or less, suggesting the importance of conducting long-duration studies given the characteristics of type 2 diabetes as a chronic disease.
Collapse
Affiliation(s)
- Manuel Ruz
- Department of Nutrition, Faculty of Medicine, University of Chile, Independencia 1027, Correo 7, Santiago, Chile.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lee SH, Kang SM, Ko SC, Kang MC, Jeon YJ. Octaphlorethol A, a novel phenolic compound isolated from Ishige foliacea, protects against streptozotocin-induced pancreatic β cell damage by reducing oxidative stress and apoptosis. Food Chem Toxicol 2013; 59:643-9. [PMID: 23871829 DOI: 10.1016/j.fct.2013.07.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/26/2013] [Accepted: 07/07/2013] [Indexed: 02/02/2023]
Abstract
Pancreatic β cells are extremely sensitive to oxidative stress, which probably has an important role in β cell damage in diabetes. The protective effect of octaphlorethol A (OPA), a novel phenolic compound isolated from Ishige foliacea, against streptozotocin (STZ)-induced pancreatic β cell damage was investigated using a rat insulinoma cell line (RINm5F pancreatic β cells). Pretreatment with OPA decreased the death of STZ-treated pancreatic β cells at concentrations of 12.5 μg/ml or 50 μg/ml, and reduced the generation of thiobarbituric acid reactive substances and intracellular reactive oxygen species in a dose-dependent manner in STZ-treated pancreatic β cells. In addition, the OPA pretreatment increased the activities of antioxidant enzymes such as catalase, superoxide dismutase, and glutathione peroxidase in STZ-treated pancreatic β cells. Moreover, OPA treatment elevated the level of insulin, which was reduced by STZ treatment, and protected pancreatic β cells against damage under STZ-treated conditions. These effects were mediated by suppressing apoptosis and were associated with increased anti-apoptotic Bcl-xL expression and reduced pro-apoptotic Bax and cleaved caspase-3 expression. These findings indicate that OPA may be useful as a potential pharmaceutical agent to protect against pancreatic β cell damage caused by oxidative stress associated with diabetes.
Collapse
Affiliation(s)
- Seung-Hong Lee
- Division of Food Bioscience, Konkuk University, Chungju Chungbuk 380-701, Republic of Korea
| | | | | | | | | |
Collapse
|
43
|
Miao X, Sun W, Fu Y, Miao L, Cai L. Zinc homeostasis in the metabolic syndrome and diabetes. Front Med 2013; 7:31-52. [PMID: 23385610 DOI: 10.1007/s11684-013-0251-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 12/26/2012] [Indexed: 12/16/2022]
Abstract
Zinc (Zn) is an essential mineral that is required for various cellular functions. Zn dyshomeostasis always is related to certain disorders such as metabolic syndrome, diabetes and diabetic complications. The associations of Zn with metabolic syndrome, diabetes and diabetic complications, thus, stem from the multiple roles of Zn: (1) a constructive component of many important enzymes or proteins, (2) a requirement for insulin storage and secretion, (3) a direct or indirect antioxidant action, and (4) an insulin-like action. However, whether there is a clear cause-and-effect relationship of Zn with metabolic syndrome, diabetes, or diabetic complications remains unclear. In fact, it is known that Zn deficiency is a common phenomenon in diabetic patients. Chronic low intake of Zn was associated with the increased risk of diabetes and diabetes also impairs Zn metabolism. Theoretically Zn supplementation should prevent the metabolic syndrome, diabetes, and diabetic complications; however, limited available data are not always supportive of the above notion. Therefore, this review has tried to summarize these pieces of available information, possible mechanisms by which Zn prevents the metabolic syndrome, diabetes, and diabetic complications. In the final part, what are the current issues for Zn supplementation were also discussed.
Collapse
Affiliation(s)
- Xiao Miao
- The Second Hospital of Jilin University, Changchun, 130021, China
| | | | | | | | | |
Collapse
|
44
|
Abstract
Zn is an essential trace element, involved in many different cellular processes. A relationship between Zn, pancreatic function and diabetes was suggested almost 70 years ago. To emphasise the importance of Zn in biology, the history of Zn research in the field of diabetes along with a general description of Zn transporter families will be reviewed. The paper will then focus on the effects of Zn on pancreatic β-cell function, including insulin synthesis and secretion, Zn signalling in the pancreatic islet, the redox functions of Zn and its target genes. The recent association of two ‘Zn genes’, i.e. metallothionein (MT) and Zn transporter 8 (SLC 30A8), with type 2 diabetes at the genetic level and with insulin secretion in clinical studies offers a potential new way to identify new drug targets to modulate Zn homeostasis directly in β-cells. The action of Zn for insulin action in its target organs, as Zn signalling in other pancreatic islet cells, will be addressed. Therapeutic Zn–insulin preparations and the influence of Zn and Zn transporters in type 1 diabetes will also be discussed. An extensive review of the literature on the clinical studies using Zn supplementation in the prevention and treatment of both types of diabetes, including complications of the disease, will evaluate the overall beneficial effects of Zn supplementation on blood glucose control, suggesting that Zn might be a candidate ion for diabetes prevention and therapy. Clearly, the story of the links between Zn, pancreatic islet cells and diabetes is only now unfolding, and we are presently only at the first chapter.
Collapse
|
45
|
Haynes V, Connor T, Tchernof A, Vidal H, Dubois S. Metallothionein 2a gene expression is increased in subcutaneous adipose tissue of type 2 diabetic patients. Mol Genet Metab 2013; 108:90-4. [PMID: 23148893 DOI: 10.1016/j.ymgme.2012.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 10/14/2012] [Accepted: 10/14/2012] [Indexed: 10/27/2022]
Abstract
STUDY BACKGROUND Insulin resistance plays an important role in the pathogenesis of type 2 diabetes and the metabolic syndrome. Many of the genes and pathways involved have been identified but some remain to be defined. Metallothioneins (Mts) are a family of anti-oxidant proteins and metallothionein 2a (Mt2a) polymorphims have been recently associated with type 2 diabetes and related complications. Our objective was to determine the Mt2a gene expression levels in adipose tissues from diabetic patients and the effect of Mt treatment on adipocyte insulin sensitivity. METHODS Samples of subcutaneous and visceral adipose tissues from lean, type 2 diabetic and non-diabetic obese patients were analysed using RT-qPCR for Mt2a mRNA abundance. The regulation of Mt2a expression was further studied in 3T3-L1 adipocytes treated or not with TNFα (10 ng/ml, 72 h) to induce insulin resistance. The effects of Mt on glucose uptake were investigated in cultured adipocytes treated with recombinant Mt protein. RESULTS We found that the Mt2a gene expression was significantly higher in adipose tissue of type 2 diabetic patients in comparison to that of lean (p=0.003) subjects. In 3T3-L1 adipocytes, insulin resistance induced by TNFα increased Mt2a mRNA levels (p=3×10(-4)) and insulin-stimulated glucose uptake was significantly inhibited by 53% (p=8×10(-4)) compared to vehicle, when 3T3-L1 adipocytes were treated with Mt protein. CONCLUSIONS These data suggest that Mt2a might be involved in insulin resistance through the up-regulation of Mt gene expression, which may lead to the modulation of insulin action in fat cells. These results suggest the concept of considering Mt proteins as markers and potential targets in type 2 diabetes.
Collapse
Affiliation(s)
- Vanessa Haynes
- Metabolic Research Unit, Deakin University, Geelong, Australia
| | | | | | | | | |
Collapse
|
46
|
Sekiguchi Y, Owada J, Oishi H, Katsumata T, Ikeda K, Kudo T, Takahashi S. Noninvasive monitoring of β-cell mass and fetal β-cell genesis in mice using bioluminescence imaging. Exp Anim 2012; 61:445-51. [PMID: 22850644 DOI: 10.1538/expanim.61.445] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Bioluminescence imaging (BLI) has been applied in gene therapy and research to screen for transgene expression, progression of infection, tumor growth and metastasis, and transplantation. It enables real-time and relatively noninvasive localization and serial quantification of biological processes in experimental animals. In diabetes research, BLI has been employed for the quantification of β-cell mass, monitoring of islet graft survival after transplantation, and detection of reporter gene expression. Here, we explore the use of BLI in a transgenic mouse expressing luciferase under the control of the mouse insulin 1 promoter (MIP-Luc-VU). A previous report on MIP-Luc-VU mice showed luminescence intensities emitted from the islets correlated well with the number of islets in vitro and in vivo. In this study, we showed MIP-Luc-VU mice fed a high fat diet for 8 weeks gave rise to a greater bioluminescent signal than mice fed a regular diet for the same period of time. Conversely, there was a strong reduction in the signal observed in diabetic Mafa-deficient/Mafk-transgenic mutant mice and streptozotocin-treated mice, reflecting the loss of β-cells. Furthermore, we were able to monitor fetal β-cell genesis in MIP-Luc-VU mice during the late gestational stage in a noninvasive and repetitive manner. In summary, we show that bioluminescence imaging of mice expressing a β-cell specific reporter allows detection of changes in β-cell mass and visualization of fetal β-cell neogenesis in uteri.
Collapse
Affiliation(s)
- Yukari Sekiguchi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1–1–1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Ergaz Z, Shoshani-Dror D, Guillemin C, Neeman-azulay M, Fudim L, Weksler-Zangen S, Stodgell CJ, Miller RK, Ornoy A. The effect of copper deficiency on fetal growth and liver anti-oxidant capacity in the Cohen diabetic rat model. Toxicol Appl Pharmacol 2012; 265:209-20. [DOI: 10.1016/j.taap.2012.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 10/03/2012] [Accepted: 10/05/2012] [Indexed: 12/20/2022]
|
48
|
Bensellam M, Laybutt DR, Jonas JC. The molecular mechanisms of pancreatic β-cell glucotoxicity: recent findings and future research directions. Mol Cell Endocrinol 2012; 364:1-27. [PMID: 22885162 DOI: 10.1016/j.mce.2012.08.003] [Citation(s) in RCA: 219] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 07/11/2012] [Accepted: 08/01/2012] [Indexed: 02/06/2023]
Abstract
It is well established that regular physiological stimulation by glucose plays a crucial role in the maintenance of the β-cell differentiated phenotype. In contrast, prolonged or repeated exposure to elevated glucose concentrations both in vitro and in vivo exerts deleterious or toxic effects on the β-cell phenotype, a concept termed as glucotoxicity. Evidence indicates that the latter may greatly contribute to the pathogenesis of type 2 diabetes. Through the activation of several mechanisms and signaling pathways, high glucose levels exert deleterious effects on β-cell function and survival and thereby, lead to the worsening of the disease over time. While the role of high glucose-induced β-cell overstimulation, oxidative stress, excessive Unfolded Protein Response (UPR) activation, and loss of differentiation in the alteration of the β-cell phenotype is well ascertained, at least in vitro and in animal models of type 2 diabetes, the role of other mechanisms such as inflammation, O-GlcNacylation, PKC activation, and amyloidogenesis requires further confirmation. On the other hand, protein glycation is an emerging mechanism that may play an important role in the glucotoxic deterioration of the β-cell phenotype. Finally, our recent evidence suggests that hypoxia may also be a new mechanism of β-cell glucotoxicity. Deciphering these molecular mechanisms of β-cell glucotoxicity is a mandatory first step toward the development of therapeutic strategies to protect β-cells and improve the functional β-cell mass in type 2 diabetes.
Collapse
Affiliation(s)
- Mohammed Bensellam
- Université catholique de Louvain, Institut de recherche expérimentale et clinique, Pôle d'endocrinologie, diabète et nutrition, Brussels, Belgium
| | | | | |
Collapse
|
49
|
Ganasyam SR, Rao TB, Murthy YSR, Jyothy A, Sujatha M. Association of Estrogen Receptor-α Gene & Metallothionein-1 Gene Polymorphisms in Type 2 Diabetic Women of Andhra Pradesh. Indian J Clin Biochem 2012; 27:69-73. [PMID: 23277715 DOI: 10.1007/s12291-011-0179-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 11/10/2011] [Indexed: 11/26/2022]
Abstract
Type 2 diabetes mellitus (DM) is a multifactorial disease where both genetic and environmental factors contribute to its pathogenesis. Estrogen plays an important role in type 2 DM pathogenesis. A number of polymorphisms have been reported in the estrogen receptor (ESR1), including the XbaI and PvuII restriction enzyme polymorphisms of ESR1,which may be involved in disease pathogenesis. Metallothioneins (MT) act as potent antioxidants against various oxidative damages. Very few studies have indicated the association between Estrogen Receptor-α, MT1 gene polymorphisms with type2 DM. A total of 100 type 2 diabetic women and 100 age, sex matched controls were recruited. Using the PCR based RFLP method, the PvuII and XbaI polymorphisms of ESR1 and in MT1A (rs8052394 and rs11076161) gene polymorphisms were analysed. The genotype distribution and frequency of mutated allele showed no significant differences between diabetic and non-diabetic groups in PvuII (χ2 = 2.443; P = 0.1181) or XbaI (χ2 = 1.789; P = 0.1812) and rs8052394 (χ2 = 1.154; P = 0.2840) or rs11076161 (χ2 = 0.4141; P = 0.5199), polymorphisms. This is the first Indian study to conclude that ESR1 and MT1 gene polymorphisms are not associated with increased susceptibility to type 2 diabetes in Indian women.
Collapse
Affiliation(s)
- Shilpa Reddy Ganasyam
- Department of Biochemistry, Institute of Genetics and Hospital for Genetic Diseases, Osmania University, Begumpet, Hyderabad, 500016 Andhra Pradesh India
| | | | | | | | | |
Collapse
|
50
|
Sun CD, Zhang B, Zhang JK, Xu CJ, Wu YL, Li X, Chen KS. Cyanidin-3-glucoside-rich extract from Chinese bayberry fruit protects pancreatic β cells and ameliorates hyperglycemia in streptozotocin-induced diabetic mice. J Med Food 2011; 15:288-98. [PMID: 22181073 DOI: 10.1089/jmf.2011.1806] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Chinese bayberry fruit is a rich source of anthocyanins, especially cyanidin-3-glucoside (C3G). The present study investigated the protective effects of C3G-rich bayberry fruit extract (CRBFE) against pancreatic β cells against oxidative stress-induced injury as well as its hypoglycemic effect in diabetic mice. Bayberry extract from "Biqi" was used for both in vitro and in vivo testing because of its high C3G content and high antioxidant capacity. Pretreatment of β cells with CRBFE (containing 0.5 μmol/L C3G) prevented cell death, increased cellular viability, and decreased mitochondrial reactive oxygen species production and cell necrosis induced by 800 or 1,200 μmol/L H₂O₂. CRBFE dose-dependently up-regulated pancreatic duodenal homeobox 1 gene expression, contributing to increased insulin-like growth factor II gene transcript levels and insulin protein in INS-1 cells. In addition, administration of CRBFE (150 μg of C3G/10 g of body weight twice per day) significantly reduced blood glucose in streptozotocin-induced diabetic ICR mice and increased the glucose tolerance in an oral glucose tolerance test (P<.05). Such results indicated that CRBFE might be useful in prevention and control of diabetes mellitus and diabetes-associated complications.
Collapse
Affiliation(s)
- Chong-De Sun
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Zijingang Campus, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|