51
|
LaPierre MP, Stoffel M. MicroRNAs as stress regulators in pancreatic beta cells and diabetes. Mol Metab 2017; 6:1010-1023. [PMID: 28951825 PMCID: PMC5605735 DOI: 10.1016/j.molmet.2017.06.020] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/29/2017] [Accepted: 06/02/2017] [Indexed: 12/12/2022] Open
Abstract
Background MicroRNAs have emerged as important regulatory non-coding RNAs that tune cellular responses to physiological perturbations and disease conditions. An increasing body of literature underlines the important roles of miRNA function in pancreatic β-cells in response to metabolic, genetic and inflammatory stress. Lessons from genetic loss- and gain-of-function studies have implicated several highly expressed and evolutionary conserved miRNAs in stress signal modulation, resolution and buffering, thereby forming stabilizing miRNA networks that preserve β-cell differentiation, function, proliferation and cell survival. Scope of Review This review will summarize our current knowledge of how biologically relevant miRNAs regulate stress responses in pancreatic β-cells, discuss the challenges and opportunities associated with using secreted miRNAs as biomarkers and forecast how mechanistic knowledge of miRNA function can be exploited in developing miRNA-based therapeutics. Major Conclusions miRNAs play important roles in the function, differentiation, proliferation, and survival of pancreatic β-cells. Many miRNA families that are regulated by metabolic, genetic, and inflammatory stressors have been found to coordinate the adaptive responses of β-cells in vivo in conditions such as obesity and diabetes.
Collapse
Affiliation(s)
| | - Markus Stoffel
- Corresponding author. Swiss Federal Institute of Technology, ETH Zürich, Institute for Molecular Health Science, Laboratory for Metabolic Diseases, Otto-Stern Weg 7, HPL H36, CH 8093 Zürich, Switzerland. Fax: +41 44 633 1362.Federal Institute of TechnologyETH ZürichInstitute for Molecular Health ScienceLaboratory for Metabolic DiseasesOtto-Stern Weg 7HPL H36ZürichCH 8093Switzerland
| |
Collapse
|
52
|
Golson ML, Kaestner KH. Epigenetics in formation, function, and failure of the endocrine pancreas. Mol Metab 2017; 6:1066-1076. [PMID: 28951829 PMCID: PMC5605720 DOI: 10.1016/j.molmet.2017.05.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/16/2017] [Accepted: 05/22/2017] [Indexed: 01/17/2023] Open
Abstract
Background Epigenetics, in the broadest sense, governs all aspects of the life of any multicellular organism, as it controls how differentiated cells arrive at their unique phenotype during development and differentiation, despite having a uniform (with some exceptions such as T-cells and germ cells) genetic make-up. The endocrine pancreas is no exception. Transcriptional regulators and epigenetic modifiers shape the differentiation of the five major endocrine cell types from their common precursor in the fetal pancreatic bud. Beyond their role in cell differentiation, interactions of the organism with the environment are also often encoded into permanent or semi-permanent epigenetic marks and affect cellular behavior and organismal health. Epigenetics is defined as any heritable – at least through one mitotic cell division – change in phenotype or trait that is not the result of a change in genomic DNA sequence, and it forms the basis that mediates the environmental impact on diabetes susceptibility and islet function. Scope of review We will summarize the impact of epigenetic regulation on islet cell development, maturation, function, and pathophysiology. We will briefly recapitulate the major epigenetic marks and their relationship to gene activity, and outline novel strategies to employ targeted epigenetic modifications as a tool to improve islet cell function. Major conclusions The improved understanding of the epigenetic underpinnings of islet cell differentiation, function and breakdown, as well as the development of innovative tools for their manipulation, is key to islet cell biology and the discovery of novel approaches to therapies for islet cell failure.
Collapse
Affiliation(s)
- Maria L Golson
- University of Pennsylvania, Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Philadelphia, PA, USA
| | - Klaus H Kaestner
- University of Pennsylvania, Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Philadelphia, PA, USA
| |
Collapse
|
53
|
Role of MicroRNAs in Type 2 Diabetes and Associated Vascular Complications. Biochimie 2017; 139:9-19. [PMID: 28487136 DOI: 10.1016/j.biochi.2017.05.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/05/2017] [Indexed: 12/20/2022]
Abstract
Type 2 diabetes mellitus (T2DM) has become a major health threat worldwide. MicroRNAs (miRNAs) are a group of non-coding RNAs known to regulate various biological processes including the pathogenesis of T2DM. Recent studies have pointed out that specific miRNAs play a critical role in controlling β cell activities and the development of diabetic vascular complications. Their association with the disease pathogenesis and omnipresence in body fluids have made them important players for prognosis, diagnosis and management of T2DM. Owing to the limitations of classical biomarkers of diabetes such as fasting plasma glucose, glycosylated haemoglobin (HbA1c) lack in predicting the risk of development of diabetes complications in a susceptible population. The miRNAs can act as ideal biomarkers for diabetes associated complications. Identification of specific miRNA signatures to detect diabetes and ideally to find out the risk of development of diabetes-associated complications in susceptible population is the essential requirement of the present clinical strategies for controlling diabetes worldwide. In this article, we summarize the potential miRNAs and miRNA signatures involved in the β cell activities and diabetes associated macrovascular and microvascular complications.
Collapse
|
54
|
Bai C, Gao Y, Li X, Wang K, Xiong H, Shan Z, Zhang P, Wang W, Guan W, Ma Y. MicroRNAs can effectively induce formation of insulin-producing cells from mesenchymal stem cells. J Tissue Eng Regen Med 2017; 11:3457-3468. [DOI: 10.1002/term.2259] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 03/28/2016] [Accepted: 07/03/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Chunyu Bai
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Yuhua Gao
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Xiangchen Li
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Kunfu Wang
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Hui Xiong
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Zhiqiang Shan
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Ping Zhang
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Wenjie Wang
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Weijun Guan
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Yuehui Ma
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| |
Collapse
|
55
|
Zheng Y, Wang Z, Zhou Z. miRNAs: novel regulators of autoimmunity-mediated pancreatic β-cell destruction in type 1 diabetes. Cell Mol Immunol 2017; 14:488-496. [PMID: 28317889 DOI: 10.1038/cmi.2017.7] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/06/2017] [Accepted: 01/06/2017] [Indexed: 02/08/2023] Open
Abstract
MicroRNAs (miRNAs) are a series of conserved, short, non-coding RNAs that modulate gene expression in a posttranscriptional manner. miRNAs are involved in almost every physiological and pathological process. Type 1 diabetes (T1D) is an autoimmune disease that is the result of selective destruction of pancreatic β-cells driven by the immune system. miRNAs are also important participants in T1D pathogenesis. Herein, we review the most recent data on the potential involvement of miRNAs in T1D. Specifically, we focus on two aspects: the roles of miRNAs in maintaining immune homeostasis and regulating β-cell survival and/or functions in T1D. We also discuss circulating miRNAs as potent biomarkers for the diagnosis and prediction of T1D and investigate potential therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Ying Zheng
- Center for Medical Research, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zhen Wang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Central South University, Changsha, Hunan 410011, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
56
|
Sebastiani G, Valentini M, Grieco GE, Ventriglia G, Nigi L, Mancarella F, Pellegrini S, Martino G, Sordi V, Piemonti L, Dotta F. MicroRNA expression profiles of human iPSCs differentiation into insulin-producing cells. Acta Diabetol 2017; 54:265-281. [PMID: 28039581 DOI: 10.1007/s00592-016-0955-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/15/2016] [Indexed: 01/10/2023]
Abstract
AIMS MicroRNAs are a class of small noncoding RNAs, which control gene expression by inhibition of mRNA translation. MicroRNAs are involved in the control of biological processes including cell differentiation. Here, we aim at characterizing microRNA expression profiles during differentiation of human induced pluripotent stem cells (hiPSCs) into insulin-producing cells. METHODS We differentiated hiPSCs toward endocrine pancreatic lineage following a 18-day protocol. We analyzed genes and microRNA expression levels using RT real-time PCR and TaqMan microRNA arrays followed by bioinformatic functional analysis. RESULTS MicroRNA expression profiles analysis of undifferentiated hiPSCs during pancreatic differentiation revealed that 347/768 microRNAs were expressed at least in one time point of all samples. We observed 18 microRNAs differentially expressed: 11 were upregulated (miR-9-5p, miR-9-3p, miR-10a, miR-99a-3p, miR-124a, miR-135a, miR-138, miR-149, miR-211, miR-342-3p and miR-375) and 7 downregulated (miR-31, miR-127, miR-143, miR-302c-3p, miR-373, miR-518b and miR-520c-3p) during differentiation into insulin-producing cells. Selected microRNAs were further evaluated during differentiation of Sendai-virus-reprogrammed hiPSCs using an improved endocrine pancreatic beta cell derivation protocol and, moreover, in differentiated NKX6.1+ sorted cells. Following Targetscan7.0 analysis of target genes of differentially expressed microRNAs and gene ontology classification, we found that such target genes belong to categories of major significance in pancreas organogenesis and development or exocytosis. CONCLUSIONS We detected a specific hiPSCs microRNAs signature during differentiation into insulin-producing cells and demonstrated that differentially expressed microRNAs target several genes involved in pancreas organogenesis.
Collapse
Affiliation(s)
- Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy
| | - Marco Valentini
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy
| | - Giuseppina Emanuela Grieco
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy
| | - Giuliana Ventriglia
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy
| | - Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy
| | - Francesca Mancarella
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy
| | - Silvia Pellegrini
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianvito Martino
- Division of Neuroscience, Institute of Experimental Neurology (INSpe), IRCCS San Raffaele Hospital, Milan, Italy
| | - Valeria Sordi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy.
| |
Collapse
|
57
|
Størling J, Pociot F. Type 1 Diabetes Candidate Genes Linked to Pancreatic Islet Cell Inflammation and Beta-Cell Apoptosis. Genes (Basel) 2017; 8:genes8020072. [PMID: 28212332 PMCID: PMC5333061 DOI: 10.3390/genes8020072] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 02/07/2017] [Accepted: 02/10/2017] [Indexed: 02/07/2023] Open
Abstract
Type 1 diabetes (T1D) is a chronic immune-mediated disease resulting from the selective destruction of the insulin-producing pancreatic islet β-cells. Susceptibility to the disease is the result of complex interactions between environmental and genetic risk factors. Genome-wide association studies (GWAS) have identified more than 50 genetic regions that affect the risk of developing T1D. Most of these susceptibility loci, however, harbor several genes, and the causal variant(s) and gene(s) for most of the loci remain to be established. A significant part of the genes located in the T1D susceptibility loci are expressed in human islets and β cells and mounting evidence suggests that some of these genes modulate the β-cell response to the immune system and viral infection and regulate apoptotic β-cell death. Here, we discuss the current status of T1D susceptibility loci and candidate genes with focus on pancreatic islet cell inflammation and β-cell apoptosis.
Collapse
Affiliation(s)
- Joachim Størling
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, University Hospital Herlev and Gentofte, Herlev 2730, Denmark.
| | - Flemming Pociot
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, University Hospital Herlev and Gentofte, Herlev 2730, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
58
|
Vienberg S, Geiger J, Madsen S, Dalgaard LT. MicroRNAs in metabolism. Acta Physiol (Oxf) 2017; 219:346-361. [PMID: 27009502 PMCID: PMC5297868 DOI: 10.1111/apha.12681] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/06/2016] [Accepted: 03/21/2016] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) have within the past decade emerged as key regulators of metabolic homoeostasis. Major tissues in intermediary metabolism important during development of the metabolic syndrome, such as β-cells, liver, skeletal and heart muscle as well as adipose tissue, have all been shown to be affected by miRNAs. In the pancreatic β-cell, a number of miRNAs are important in maintaining the balance between differentiation and proliferation (miR-200 and miR-29 families) and insulin exocytosis in the differentiated state is controlled by miR-7, miR-375 and miR-335. MiR-33a and MiR-33b play crucial roles in cholesterol and lipid metabolism, whereas miR-103 and miR-107 regulates hepatic insulin sensitivity. In muscle tissue, a defined number of miRNAs (miR-1, miR-133, miR-206) control myofibre type switch and induce myogenic differentiation programmes. Similarly, in adipose tissue, a defined number of miRNAs control white to brown adipocyte conversion or differentiation (miR-365, miR-133, miR-455). The discovery of circulating miRNAs in exosomes emphasizes their importance as both endocrine signalling molecules and potentially disease markers. Their dysregulation in metabolic diseases, such as obesity, type 2 diabetes and atherosclerosis stresses their potential as therapeutic targets. This review emphasizes current ideas and controversies within miRNA research in metabolism.
Collapse
Affiliation(s)
- S. Vienberg
- Center for Basic Metabolic ResearchFaculty of HealthUniversity of CopenhagenCopenhagenDenmark
| | - J. Geiger
- Department of Science and EnvironmentRoskilde UniversityRoskildeDenmark
| | - S. Madsen
- Center for Basic Metabolic ResearchFaculty of HealthUniversity of CopenhagenCopenhagenDenmark
| | - L. T. Dalgaard
- Department of Science and EnvironmentRoskilde UniversityRoskildeDenmark
| |
Collapse
|
59
|
Eliasson L, Esguerra JLS, Wendt A. Lessons from basic pancreatic beta cell research in type-2 diabetes and vascular complications. Diabetol Int 2017; 8:139-152. [PMID: 30603317 DOI: 10.1007/s13340-017-0304-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/08/2017] [Indexed: 12/14/2022]
Abstract
The changes in life-style with increased access of food and reduced physical activity have resulted in the global epidemic of obesity. Consequently, individuals with type 2 diabetes and cardiovascular disease have also escalated. A central organ in the development of diabetes is the pancreas, and more specifically the pancreatic beta cells within the islets of Langerhans. Beta cells have been assigned the important task of secreting insulin when blood glucose is increased to lower the glucose level. An early sign of diabetes pathogenesis is lack of first phase insulin response and reduced second phase secretion. In this review, which is based on the foreign investigator award lecture given at the JSDC meeting in Sendai in October 2016, we discuss a possible cellular explanation for the reduced first phase insulin response and how this can be influenced by lipids. Moreover, since patients with cardiovascular disease and high levels of cholesterol are often treated with statins, we summarize recent data regarding effects on statins on glucose homeostasis and insulin secretion. Finally, we suggest microRNAs (miRNAs) as central players in the adjustment of beta cell function during the development of diabetes. We specifically discuss miRNAs regarding their involvement in insulin secretion regulation, differential expression in type 2 diabetes, and potential as biomarkers for prediction of diabetes and cardiovascular complications.
Collapse
Affiliation(s)
- Lena Eliasson
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| | - Jonathan Lou S Esguerra
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| | - Anna Wendt
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| |
Collapse
|
60
|
Dalgaard LT, Eliasson L. An 'alpha-beta' of pancreatic islet microribonucleotides. Int J Biochem Cell Biol 2017; 88:208-219. [PMID: 28122254 DOI: 10.1016/j.biocel.2017.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 11/17/2022]
Abstract
MicroRNAs (miRNAs) are cellular, short, non-coding ribonucleotides acting as endogenous posttranscriptional repressors following incorporation in the RNA-induced silencing complex. Despite being chemically and mechanistically very similar, miRNAs exert a multitude of different cellular effects by acting on mRNA species, whose gene-products partake in a wide array of processes. Here, the aim was to review the knowledge of miRNA expression and action in the islet of Langerhans. We have focused on: 1) physiological consequences of islet or beta cell specific inhibition of miRNA processing, 2) mechanisms regulating processing of miRNAs in islet cells, 3) presence and function of miRNAs in alpha versus beta cells - the two main cell types of islets, and 4) miRNA mediators of beta cell decompensation. It is clear that miRNAs regulate pancreatic islet development, maturation, and function in vivo. Moreover, processing of miRNAs appears to be altered by obesity, diabetes, and aging. A number of miRNAs (such as miR-7, miR-21, miR-29, miR-34a, miR-212/miR-132, miR-184, miR-200 and miR-375) are involved in mediating beta cell dysfunction and/or compensation induced by hyperglycemia, oxidative stress, cytotoxic cytokines, and in rodent models of fetal metabolic programming prediabetes and overt diabetes. Studies of human type 2 diabetic islets underline that these miRNA families could have important roles also in human type 2 diabetes. Furthermore, there is a genuine gap of knowledge regarding miRNA expression and function in pancreatic alpha cells. Progress in this area would be enhanced by improved in vitro alpha cell models and better tools for islet cell sorting.
Collapse
Affiliation(s)
| | - Lena Eliasson
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, CRC, SUS, Malmö, Sweden.
| |
Collapse
|
61
|
Martinez-Sanchez A, Rutter GA, Latreille M. MiRNAs in β-Cell Development, Identity, and Disease. Front Genet 2017; 7:226. [PMID: 28123396 PMCID: PMC5225124 DOI: 10.3389/fgene.2016.00226] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/21/2016] [Indexed: 12/22/2022] Open
Abstract
Pancreatic β-cells regulate glucose metabolism by secreting insulin, which in turn stimulates the utilization or storage of the sugar by peripheral tissues. Insulin insufficiency and a prolonged period of insulin resistance are usually the core components of type 2 diabetes (T2D). Although, decreased insulin levels in T2D have long been attributed to a decrease in β-cell function and/or mass, this model has recently been refined with the recognition that a loss of β-cell “identity” and dedifferentiation also contribute to the decline in insulin production. MicroRNAs (miRNAs) are key regulatory molecules that display tissue-specific expression patterns and maintain the differentiated state of somatic cells. During the past few years, great strides have been made in understanding how miRNA circuits impact β-cell identity. Here, we review current knowledge on the role of miRNAs in regulating the acquisition of the β-cell fate during development and in maintaining mature β-cell identity and function during stress situations such as obesity, pregnancy, aging, or diabetes. We also discuss how miRNA function could be harnessed to improve our ability to generate β-cells for replacement therapy for T2D.
Collapse
Affiliation(s)
- Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London London, UK
| | - Mathieu Latreille
- Cellular Identity and Metabolism Group, MRC London Institute of Medical SciencesLondon, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College LondonLondon, UK
| |
Collapse
|
62
|
Bernstein D, Golson ML, Kaestner KH. Epigenetic control of β-cell function and failure. Diabetes Res Clin Pract 2017; 123:24-36. [PMID: 27918975 PMCID: PMC5250585 DOI: 10.1016/j.diabres.2016.11.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022]
Abstract
Type 2 diabetes is a highly heritable disease, but only ∼15% of this heritability can be explained by known genetic variant loci. In fact, body mass index is more predictive of diabetes than any of the common risk alleles identified by genome-wide association studies. This discrepancy may be explained by epigenetic inheritance, whereby changes in gene regulation can be passed along to offspring. Epigenetic changes throughout an organism's lifetime, based on environmental factors such as chemical exposures, diet, physical activity, and age, can also affect gene expression and susceptibility to diabetes. Recently, novel genome-wide assays of epigenetic marks have resulted in a greater understanding of how genetics, epigenetics, and the environment interact in the development and inheritance of diabetes.
Collapse
Affiliation(s)
- Diana Bernstein
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria L Golson
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
63
|
Dumortier O, Fabris G, Van Obberghen E. Shaping and preserving β-cell identity with microRNAs. Diabetes Obes Metab 2016; 18 Suppl 1:51-7. [PMID: 27615131 DOI: 10.1111/dom.12722] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/26/2016] [Indexed: 12/18/2022]
Abstract
The highly sophisticated identity of pancreatic β-cells is geared to accomplish its unique feat of providing insulin for organismal glucose and lipid homeostasis. This requires a particular and streamlined fuel metabolism which defines mature β-cells as glucose sensors linked to an insulin exocytosis machinery. The establishment of an appropriate β-cell mass and function during development as well as the maintenance of their identity throughout life are necessary for energy homeostasis. The small non-coding RNAs, microRNAs (miRNAs), are now well-recognized regulators of gene transcripts, which in general are negatively affected by them. Convincing evidence exists to view miRNAs as major actors in β-cell development and function, suggesting an important role for them in the distinctive β-cell 'identity card'. Here, we summarize key features that associate miRNAs and the establishment of the appropriate β-cell identity and its necessary maintenance during their 'long life'.
Collapse
Affiliation(s)
- O Dumortier
- University Côte d'Azur, Inserm, CNRS, IRCAN, France
| | - G Fabris
- University Côte d'Azur, Inserm, CNRS, IRCAN, France
| | | |
Collapse
|
64
|
Osmai M, Osmai Y, Bang-Berthelsen CH, Pallesen EMH, Vestergaard AL, Novotny GW, Pociot F, Mandrup-Poulsen T. MicroRNAs as regulators of beta-cell function and dysfunction. Diabetes Metab Res Rev 2016; 32:334-49. [PMID: 26418758 DOI: 10.1002/dmrr.2719] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/21/2015] [Accepted: 08/13/2015] [Indexed: 12/14/2022]
Abstract
In the last decade, there has been an explosion in both the number of and knowledge about miRNAs associated with both type 1 and type 2 diabetes. Even though we are presently in the initial stages of understanding how this novel class of posttranscriptional regulators are involved in diabetes, recent studies have demonstrated that miRNAs are important regulators of the islet transcriptome, controlling apoptosis, differentiation and proliferation, as well as regulating unique islet and beta-cell functions and pathways such as insulin expression, processing and secretion. Furthermore, a large number of miRNAs have been linked to diabetogenic processes induced by elevated levels of glucose, free fatty acids and inflammatory cytokines. Thus, miRNAs are novel therapeutic targets with the potential of protecting the beta-cell, and there is proof of principle that miRNA antagonists, so-called antagomirs, are effective in vivo for other disorders. miRNAs are exported out of cells in exosomes, raising the intriguing possibility of cell-to-cell communication between distant tissues via miRNAs and that miRNAs can be used as biomarkers of beta-cell function, mass and survival. The purpose of this review is to provide a status on how miRNAs control beta-cell function and viability in health and disease.
Collapse
Affiliation(s)
- Mirwais Osmai
- Immuno-endocrinology Lab, Section of Endocrinological Research, Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Yama Osmai
- Immuno-endocrinology Lab, Section of Endocrinological Research, Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Claus H Bang-Berthelsen
- Department of Pediatrics and Center for Non-Coding RNA in Technology and Health, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
- Diabetes NBEs and Obesity Biology, Novo Nordisk A/S, Måløv, Denmark
| | - Emil M H Pallesen
- Immuno-endocrinology Lab, Section of Endocrinological Research, Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Anna L Vestergaard
- Immuno-endocrinology Lab, Section of Endocrinological Research, Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Guy W Novotny
- Immuno-endocrinology Lab, Section of Endocrinological Research, Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Pociot
- Department of Pediatrics and Center for Non-Coding RNA in Technology and Health, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Mandrup-Poulsen
- Immuno-endocrinology Lab, Section of Endocrinological Research, Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
65
|
Malm HA, Mollet IG, Berggreen C, Orho-Melander M, Esguerra JLS, Göransson O, Eliasson L. Transcriptional regulation of the miR-212/miR-132 cluster in insulin-secreting β-cells by cAMP-regulated transcriptional co-activator 1 and salt-inducible kinases. Mol Cell Endocrinol 2016; 424:23-33. [PMID: 26797246 DOI: 10.1016/j.mce.2016.01.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 12/15/2015] [Accepted: 01/11/2016] [Indexed: 12/25/2022]
Abstract
MicroRNAs are central players in the control of insulin secretion, but their transcriptional regulation is poorly understood. Our aim was to investigate cAMP-mediated transcriptional regulation of the miR-212/miR-132 cluster and involvement of further upstream proteins in insulin secreting β-cells. cAMP induced by forskolin+IBMX or GLP-1 caused increased expression of miR-212/miR-132, and elevated phosphorylation of cAMP-response-element-binding-protein (CREB)/Activating-transcription-factor-1 (ATF1) and Salt-Inducible-Kinases (SIKs). CyclicAMP-Regulated Transcriptional Co-activator-1 (CRTC1) was concomitantly dephosphorylated and translocated to the nucleus. Silencing of miR-212/miR-132 reduced, and overexpression of miR-212 increased, glucose-stimulated insulin secretion. Silencing of CRTC1 expression resulted in decreased insulin secretion and miR-212/miR-132 expression, while silencing or inhibition of SIKs was associated with increased expression of the microRNAs and dephosphorylation of CRTC1. CRTC1 protein levels were reduced after silencing of miR-132, suggesting feed-back regulation. Our data propose cAMP-dependent co-regulation of miR-212/miR-132, in part mediated through SIK-regulated CRTC1, as an important factor for fine-tuned regulation of insulin secretion.
Collapse
Affiliation(s)
- Helena Anna Malm
- Lund University Diabetes Centre, Lund University, Unit of Islet Cell Exocytosis, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden; Lund University Diabetes Centre, Lund University, Unit of Diabetes and Cardiovascular Disease, Genetic Epidemiology, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden
| | - Inês G Mollet
- Lund University Diabetes Centre, Lund University, Unit of Islet Cell Exocytosis, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden; Lund University Diabetes Centre, Lund University, Unit of Diabetes and Cardiovascular Disease, Genetic Epidemiology, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden
| | - Christine Berggreen
- Lund University Diabetes Centre, Lund University, Protein Phosphorylation Research Unit, Dept. Experimental Medical Science, 221 84 Lund, Sweden
| | - Marju Orho-Melander
- Lund University Diabetes Centre, Lund University, Unit of Diabetes and Cardiovascular Disease, Genetic Epidemiology, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden
| | - Jonathan Lou S Esguerra
- Lund University Diabetes Centre, Lund University, Unit of Islet Cell Exocytosis, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden
| | - Olga Göransson
- Lund University Diabetes Centre, Lund University, Protein Phosphorylation Research Unit, Dept. Experimental Medical Science, 221 84 Lund, Sweden
| | - Lena Eliasson
- Lund University Diabetes Centre, Lund University, Unit of Islet Cell Exocytosis, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden.
| |
Collapse
|
66
|
Role of microRNA-21 in the formation of insulin-producing cells from pancreatic progenitor cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:280-93. [DOI: 10.1016/j.bbagrm.2015.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/17/2015] [Accepted: 12/02/2015] [Indexed: 12/20/2022]
|
67
|
DiStefano JK. Beyond the Protein-Coding Sequence: Noncoding RNAs in the Pathogenesis of Type 2 Diabetes. Rev Diabet Stud 2016; 12:260-76. [PMID: 26859655 DOI: 10.1900/rds.2015.12.260] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Diabetes mellitus results from a deficiency or failure to maintain normal glucose homeostasis. The most common form of the disease is type 2 diabetes (T2D), a progressive metabolic disorder characterized by elevated glucose levels that develops in response to either multi-organ insulin resistance or insufficient insulin secretion from pancreatic β-cells. Although the etiology of T2D is complex, many factors are known to contribute to defects of glucose homeostasis, including obesity, unhealthy lifestyle choices, genetic susceptibility, and environmental exposures. In addition to these factors, noncoding RNAs (ncRNAs) have been recently implicated in the pathogenesis of T2D, playing roles in several of the pathophysiological mechanisms underlying the disease, particularly in insulin-sensitive tissues such as pancreatic β-cells, liver, muscle, and adipose tissue. A growing number of publications demonstrate that polymorphisms in ncRNAs or their target genes may represent a new class of genetic variation contributing to the development of T2D. This review summarizes both the current state of knowledge of ncRNAs, specifically microRNAs (miRNAs), involved in the regulation of β-cell function, insulin sensitivity, and insulin action in peripheral organs. The role of genetic variation in miRNAs or miRNA binding sites in the pathogenesis of T2D is also discussed. While far less is known about the impact of long ncRNAs (lncRNAs) in the development of T2D, emerging evidence suggests that these molecules may be able to contribute to β-cell dysfunction in response to hyperglycemia. This article provides an overview of the studies conducted to date in this field, focusing on lncRNAs that are dysregulated in human pancreatic islets.
Collapse
|
68
|
Dooley J, Garcia-Perez JE, Sreenivasan J, Schlenner SM, Vangoitsenhoven R, Papadopoulou AS, Tian L, Schonefeldt S, Serneels L, Deroose C, Staats KA, Van der Schueren B, De Strooper B, McGuinness OP, Mathieu C, Liston A. The microRNA-29 Family Dictates the Balance Between Homeostatic and Pathological Glucose Handling in Diabetes and Obesity. Diabetes 2016; 65:53-61. [PMID: 26696639 PMCID: PMC4876765 DOI: 10.2337/db15-0770] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The microRNA-29 (miR-29) family is among the most abundantly expressed microRNA in the pancreas and liver. Here, we investigated the function of miR-29 in glucose regulation using miR-29a/b-1 (miR-29a)-deficient mice and newly generated miR-29b-2/c (miR-29c)-deficient mice. We observed multiple independent functions of the miR-29 family, which can be segregated into a hierarchical physiologic regulation of glucose handling. miR-29a, and not miR-29c, was observed to be a positive regulator of insulin secretion in vivo, with dysregulation of the exocytotic machinery sensitizing β-cells to overt diabetes after unfolded protein stress. By contrast, in the liver both miR-29a and miR-29c were important negative regulators of insulin signaling via phosphatidylinositol 3-kinase regulation. Global or hepatic insufficiency of miR-29 potently inhibited obesity and prevented the onset of diet-induced insulin resistance. These results demonstrate strong regulatory functions for the miR-29 family in obesity and diabetes, culminating in a hierarchical and dose-dependent effect on premature lethality.
Collapse
Affiliation(s)
- James Dooley
- VIB, Leuven, Belgium Department of Microbiology and Immunology, KUL - University of Leuven, Leuven, Belgium
| | - Josselyn E Garcia-Perez
- VIB, Leuven, Belgium Department of Microbiology and Immunology, KUL - University of Leuven, Leuven, Belgium
| | - Jayasree Sreenivasan
- VIB, Leuven, Belgium Department of Microbiology and Immunology, KUL - University of Leuven, Leuven, Belgium Department of Oncology, KUL - University of Leuven, Leuven, Belgium
| | - Susan M Schlenner
- VIB, Leuven, Belgium Department of Microbiology and Immunology, KUL - University of Leuven, Leuven, Belgium
| | - Roman Vangoitsenhoven
- Department of Clinical and Experimental Medicine, KUL - University of Leuven, Leuven, Belgium
| | | | - Lei Tian
- VIB, Leuven, Belgium Department of Microbiology and Immunology, KUL - University of Leuven, Leuven, Belgium
| | - Susann Schonefeldt
- VIB, Leuven, Belgium Department of Microbiology and Immunology, KUL - University of Leuven, Leuven, Belgium
| | - Lutgarde Serneels
- VIB, Leuven, Belgium Center for Human Genetics, KUL - University of Leuven, Leuven, Belgium
| | - Christophe Deroose
- Department of Imaging and Pathology, KUL - University of Leuven, Leuven, Belgium
| | - Kim A Staats
- VIB, Leuven, Belgium Department of Microbiology and Immunology, KUL - University of Leuven, Leuven, Belgium
| | - Bart Van der Schueren
- Department of Clinical and Experimental Medicine, KUL - University of Leuven, Leuven, Belgium
| | - Bart De Strooper
- VIB, Leuven, Belgium Center for Human Genetics, KUL - University of Leuven, Leuven, Belgium
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Chantal Mathieu
- Department of Clinical and Experimental Medicine, KUL - University of Leuven, Leuven, Belgium
| | - Adrian Liston
- VIB, Leuven, Belgium Department of Microbiology and Immunology, KUL - University of Leuven, Leuven, Belgium
| |
Collapse
|
69
|
Abstract
Pancreas development is controlled by a complex interaction of signaling pathways and transcription factor networks that determine pancreatic specification and differentiation of exocrine and endocrine cells. Epigenetics adds a new layer of gene regulation. DNA methylation, histone modifications and non-coding RNAs recently appeared as important epigenetic factors regulating pancreas development. In this review, we report recent findings obtained by analyses in model organisms as well as genome-wide approaches that demonstrate the role of these epigenetic regulators in the control of exocrine and endocrine cell differentiation, identity, function, proliferation and regeneration. We also highlight how altered epigenetic processes contribute to pancreatic disorders: diabetes and pancreatic cancer. Uncovering these epigenetic events can help to better understand these diseases, provide novel therapeutical targets for their treatment, and improve cell-based therapies for diabetes.
Collapse
Affiliation(s)
- Evans Quilichini
- Centre National de la Recherche Scientifique (CNRS), UMR7622, Institut de Biologie Paris-Seine (IBPS), Paris F-75005, France; Sorbonne Universités, UPMC Université Paris 06, UMR7622-IBPS, Paris F-75005, France
| | - Cécile Haumaitre
- Centre National de la Recherche Scientifique (CNRS), UMR7622, Institut de Biologie Paris-Seine (IBPS), Paris F-75005, France; Sorbonne Universités, UPMC Université Paris 06, UMR7622-IBPS, Paris F-75005, France; Institut National de la Santé et de la Recherche Médicale (INSERM), France.
| |
Collapse
|
70
|
Abstract
Type 1 diabetes (T1D) is a multifactorial disease resulting from an immune-mediated destruction of the insulin-producing pancreatic β cells. Several environmental and genetic risk factors predispose to the disease. Genome-wide association studies (GWAS) have identified around 50 genetic regions that affect the risk of developing T1D, but the disease-causing variants and genes are still largely unknown. In this review, we discuss the current status of T1D susceptibility loci and candidate genes with focus on the β cell. At least 40 % of the genes in the T1D susceptibility loci are expressed in human islets and β cells, where they according to recent studies modulate the β-cell response to the immune system. As most of the risk variants map to noncoding regions of the genome, i.e., promoters, enhancers, intergenic regions, and noncoding genes, their possible involvement in T1D pathogenesis as gene regulators will also be addressed.
Collapse
Affiliation(s)
- Tina Fløyel
- Copenhagen Diabetes Research Center, Department of Pediatrics, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730, Herlev, Denmark.
| | - Simranjeet Kaur
- Copenhagen Diabetes Research Center, Department of Pediatrics, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730, Herlev, Denmark.
| | - Flemming Pociot
- Copenhagen Diabetes Research Center, Department of Pediatrics, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730, Herlev, Denmark.
| |
Collapse
|
71
|
Guay C, Regazzi R. MicroRNAs and the functional β cell mass: For better or worse. DIABETES & METABOLISM 2015; 41:369-77. [DOI: 10.1016/j.diabet.2015.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/18/2015] [Accepted: 03/22/2015] [Indexed: 12/20/2022]
|
72
|
Postnatal β-cell maturation is associated with islet-specific microRNA changes induced by nutrient shifts at weaning. Nat Commun 2015; 6:8084. [PMID: 26330140 PMCID: PMC4569696 DOI: 10.1038/ncomms9084] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/16/2015] [Indexed: 12/21/2022] Open
Abstract
Glucose-induced insulin secretion is an essential function of pancreatic β-cells that is partially lost in individuals affected by Type 2 diabetes. This unique property of β-cells is acquired through a poorly understood postnatal maturation process involving major modifications in gene expression programs. Here we show that β-cell maturation is associated with changes in microRNA expression induced by the nutritional transition that occurs at weaning. When mimicked in newborn islet cells, modifications in the level of specific microRNAs result in a switch in the expression of metabolic enzymes and cause the acquisition of glucose-induced insulin release. Our data suggest microRNAs have a central role in postnatal β-cell maturation and in the determination of adult functional β-cell mass. A better understanding of the events governing β-cell maturation may help understand why some individuals are predisposed to developing diabetes and could lead to new strategies for the treatment of this common metabolic disease. Pancreatic β-cells are less responsive to changes in glucose concentration in newborn than in adult rats. Here, the authors show that functional β-cell maturation is associated with changes in miRNA expression induced by nutritional shifts at the suckling-to-weaning transition.
Collapse
|
73
|
Abstract
MicroRNAs are small noncoding ribonucleotides that regulate mRNA translation or degradation and have major roles in cellular function. MicroRNA (miRNA) levels are deregulated or altered in many diseases. There is overwhelming evidence that miRNAs also play an important role in the regulation of glucose homeostasis and thereby may contribute to the establishment of diabetes. MiRNAs have been shown to affect insulin levels by regulating insulin production, insulin exocytosis, and endocrine pancreas development. Although a large number of miRNAs have been identified from pancreatic β-cells using various screens, functional studies that link most of the identified miRNAs to regulation of pancreatic β-cell function are lacking. This review focuses on miRNAs with important roles in regulation of insulin production, insulin secretion, and β-cell development, and will discuss only miRNAs with established roles in β-cell function.
Collapse
Affiliation(s)
- Sabire Ozcan
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| |
Collapse
|
74
|
Martinez-Sanchez A, Nguyen-Tu MS, Rutter GA. DICER Inactivation Identifies Pancreatic β-Cell "Disallowed" Genes Targeted by MicroRNAs. Mol Endocrinol 2015; 29:1067-79. [PMID: 26038943 PMCID: PMC4484783 DOI: 10.1210/me.2015-1059] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pancreatic β-cells are the body's sole source of circulating insulin and essential for the maintenance of blood glucose homeostasis. Levels of up to 66 “disallowed” genes, which are strongly expressed and play housekeeping roles in most other mammalian tissues, are unusually low in β-cells. The molecular mechanisms involved in repressing these genes are largely unknown. Here, we explore the role in gene disallowance of microRNAs (miRNAs), a type of small noncoding RNAs that silence gene expression at the posttranscriptional level and are essential for β-cell development and function. To selectively deplete miRNAs from adult β-cells, the miRNA-processing enzyme DICER was inactivated by deletion of the RNase III domain with a tamoxifen-inducible Pdx1CreER transgene. In this model, β-cell dysfunction was apparent 2 weeks after recombination and preceded a decrease in insulin content and loss of β-cell mass. Of the 14 disallowed genes studied, quantitative RT-quantitative real-time PCR revealed that 6 genes (Fcgrt, Igfbp4, Maf, Oat, Pdgfra, and Slc16a1) were up-regulated (1.4- to 2.1-fold, P < .05) at this early stage. Expression of luciferase constructs bearing the 3′-untranslated regions of the corresponding mRNAs in wild-type or DICER-null β-cells demonstrated that Fcgrt, Oat, and Pdgfra are miRNA direct targets. We thus reveal a role for miRNAs in the regulation of disallowed genes in β-cells and provide evidence for a novel means through which noncoding RNAs control the functional identity of these cells independently of actions on β-cell mass.
Collapse
Affiliation(s)
- Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Marie-Sophie Nguyen-Tu
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| |
Collapse
|
75
|
Farr RJ, Januszewski AS, Joglekar MV, Liang H, McAulley AK, Hewitt AW, Thomas HE, Loudovaris T, Kay TWH, Jenkins A, Hardikar AA. A comparative analysis of high-throughput platforms for validation of a circulating microRNA signature in diabetic retinopathy. Sci Rep 2015; 5:10375. [PMID: 26035063 PMCID: PMC4649912 DOI: 10.1038/srep10375] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 04/01/2015] [Indexed: 01/02/2023] Open
Abstract
MicroRNAs are now increasingly recognized as biomarkers of disease progression. Several quantitative real-time PCR (qPCR) platforms have been developed to determine the relative levels of microRNAs in biological fluids. We systematically compared the detection of cellular and circulating microRNA using a standard 96-well platform, a high-content microfluidics platform and two ultra-high content platforms. We used extensive analytical tools to compute inter- and intra-run variability and concordance measured using fidelity scoring, coefficient of variation and cluster analysis. We carried out unprejudiced next generation sequencing to identify a microRNA signature for Diabetic Retinopathy (DR) and systematically assessed the validation of this signature on clinical samples using each of the above four qPCR platforms. The results indicate that sensitivity to measure low copy number microRNAs is inversely related to qPCR reaction volume and that the choice of platform for microRNA biomarker validation should be made based on the abundance of miRNAs of interest.
Collapse
Affiliation(s)
- Ryan J Farr
- Diabetes and Islet biology Group, NHMRC Clinical Trials Centre, Faculty of Medicine, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Andrzej S Januszewski
- Biomarkers Laboratory, NHMRC Clinical Trials Centre, Faculty of Medicine, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Mugdha V Joglekar
- Diabetes and Islet biology Group, NHMRC Clinical Trials Centre, Faculty of Medicine, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Helena Liang
- Clinical Genetics Unit, Center for Eye Research Australia (CERA), The University of Melbourne, Royal Victorian Eye and Ear Hospital, Peter Howson Wing, Level 1, 32 Gisborne Street, Melbourne, VIC 3002, Australia
| | - Annie K McAulley
- Clinical Genetics Unit, Center for Eye Research Australia (CERA), The University of Melbourne, Royal Victorian Eye and Ear Hospital, Peter Howson Wing, Level 1, 32 Gisborne Street, Melbourne, VIC 3002, Australia
| | - Alex W Hewitt
- Clinical Genetics Unit, Center for Eye Research Australia (CERA), The University of Melbourne, Royal Victorian Eye and Ear Hospital, Peter Howson Wing, Level 1, 32 Gisborne Street, Melbourne, VIC 3002, Australia
| | - Helen E Thomas
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
| | - Tom Loudovaris
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
| | - Thomas W H Kay
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
| | - Alicia Jenkins
- Biomarkers Laboratory, NHMRC Clinical Trials Centre, Faculty of Medicine, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Anandwardhan A Hardikar
- Diabetes and Islet biology Group, NHMRC Clinical Trials Centre, Faculty of Medicine, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| |
Collapse
|
76
|
Sebastiani G, Po A, Miele E, Ventriglia G, Ceccarelli E, Bugliani M, Marselli L, Marchetti P, Gulino A, Ferretti E, Dotta F. MicroRNA-124a is hyperexpressed in type 2 diabetic human pancreatic islets and negatively regulates insulin secretion. Acta Diabetol 2015; 52:523-30. [PMID: 25408296 DOI: 10.1007/s00592-014-0675-y] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/24/2014] [Indexed: 11/27/2022]
Abstract
AIMS MicroRNAs are a class of negative regulators of gene expression, which have been shown to be involved in the development of endocrine pancreas and in the regulation of insulin secretion. Since type 2 diabetes (T2D) is characterized by beta cell dysfunction, we aimed at evaluating expression levels of miR-124a and miR-375, both involved in the control of beta cell function, in human pancreatic islets obtained from T2D and from age-matched non-diabetic organ donors. METHODS We analyzed miR-124a and miR-375 expression by real-time qRT-PCR in human pancreatic islets and evaluated the potential role of miR-124a by overexpressing or silencing such miRNA in MIN6 pseudoislets. RESULTS We identified a major miR-124a hyperexpression in T2D human pancreatic islets with no differential expression of miR-375. Of note, miR-124a overexpression in MIN6 pseudoislets resulted in an impaired glucose-induced insulin secretion. In addition, miR-124a silencing in MIN6 pseudoislets resulted in increased expression of predicted target genes (Mtpn, Foxa2, Flot2, Akt3, Sirt1 and NeuroD1) involved in beta cell function. For Mtpn and Foxa2, we further demonstrated the actual binding of miR-124a to their 3UTR sequences by luciferase assay. CONCLUSIONS We uncovered a major hyperexpression of miR-124a in T2D islets, whose silencing resulted in increased expression of target genes of major importance for beta cell function and whose overexpression impaired glucose-stimulated insulin secretion, leading to the hypothesis that an altered miR-124a expression may contribute to beta cell dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- Guido Sebastiani
- Fondazione Umberto Di Mario ONLUS, c/o Toscana Life Science Park, Siena, Italy,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Mathiyalagan P, Keating ST, Al-Hasani K, El-Osta A. Epigenetic-mediated reprogramming of pancreatic endocrine cells. Antioxid Redox Signal 2015; 22:1483-95. [PMID: 25621632 DOI: 10.1089/ars.2014.6103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE Type 1 diabetes (T1D) results from cell-mediated autoimmune destruction of insulin-secreting pancreatic beta cells (β-cells). In the context of T1D, the scarcity of organ donors has driven research to alternate sources of functionally competent, insulin-secreting β-cells as substitute for donor islets to meet the clinical need for transplantation therapy. RECENT ADVANCES Experimental evidence of an inherent plasticity of pancreatic cells has fuelled interest in in vivo regeneration of β-cells. Transcriptional modulation and direct reprogramming of noninsulin secreting pancreatic α-cells to functionally mimic insulin-secreting β-cells is one of the promising avenues to the treatment of diabetes. Recent studies now show that adult progenitor and glucagon(+) α-cells can be converted into β-like cells in vivo, as a result of specific activation of the Pax4 gene in α-cells and curing diabetes in preclinical models. CRITICAL ISSUES The challenge now is to understand the precise developmental transitions mediated by endocrine transcription factors and co-regulatory determinants responsible for pancreatic function and repair. FUTURE DIRECTIONS Epigenetic-mediated regulation of transcription factor binding in pancreatic α-cells by specific drugs to direct reprogramming into functional insulin producing cells could be of potential innovative therapy for the treatment of T1D.
Collapse
Affiliation(s)
- Prabhu Mathiyalagan
- 1 Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct , Melbourne, Australia
| | | | | | | |
Collapse
|
78
|
Pancreatic β-cell identity, glucose sensing and the control of insulin secretion. Biochem J 2015; 466:203-18. [PMID: 25697093 DOI: 10.1042/bj20141384] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Insulin release from pancreatic β-cells is required to maintain normal glucose homoeostasis in man and many other animals. Defective insulin secretion underlies all forms of diabetes mellitus, a disease currently reaching epidemic proportions worldwide. Although the destruction of β-cells is responsible for Type 1 diabetes (T1D), both lowered β-cell mass and loss of secretory function are implicated in Type 2 diabetes (T2D). Emerging results suggest that a functional deficiency, involving de-differentiation of the mature β-cell towards a more progenitor-like state, may be an important driver for impaired secretion in T2D. Conversely, at least in rodents, reprogramming of islet non-β to β-cells appears to occur spontaneously in models of T1D, and may occur in man. In the present paper, we summarize the biochemical properties which define the 'identity' of the mature β-cell as a glucose sensor par excellence. In particular, we discuss the importance of suppressing a group of 11 'disallowed' housekeeping genes, including Ldha and the monocarboxylate transporter Mct1 (Slc16a1), for normal nutrient sensing. We then survey the changes in the expression and/or activity of β-cell-enriched transcription factors, including FOXO1, PDX1, NKX6.1, MAFA and RFX6, as well as non-coding RNAs, which may contribute to β-cell de-differentiation and functional impairment in T2D. The relevance of these observations for the development of new approaches to treat T1D and T2D is considered.
Collapse
|
79
|
Abstract
PURPOSE OF REVIEW The identification and characterization of essential islet transcription factors have improved our understanding of β cell development, provided insights into many of the cellular dysfunctions related to diabetes, and facilitated the successful generation of β cells from alternative cell sources. Recently, noncoding RNAs have emerged as a novel set of molecules that may represent missing components of the known islet regulatory pathways. The purpose of this article is to highlight studies that have implicated noncoding RNAs as important regulators of pancreas cell development and β cell function. RECENT FINDINGS Disruption of essential components of the microRNA processing machinery, in addition to misregulation of individual microRNAs, has revealed the importance of microRNAs in pancreas development and β cell function. Furthermore, over 1000 islet-specific long noncoding RNAs have been identified in mouse and human islets, suggesting that this class of noncoding molecules will also play important functional roles in the β cell. SUMMARY The analysis of noncoding RNAs in the pancreas will provide important new insights into pancreatic regulatory processes that will improve our ability to understand and treat diabetes, and may facilitate the generation of replacement β cells from alternative cell sources.
Collapse
Affiliation(s)
- Ruth A Singer
- Department of Genetics and Development, Columbia University, New York, New York, USA
| | | | | |
Collapse
|
80
|
Guay C, Regazzi R. Role of islet microRNAs in diabetes: which model for which question? Diabetologia 2015; 58:456-63. [PMID: 25512004 DOI: 10.1007/s00125-014-3471-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/14/2014] [Indexed: 10/24/2022]
Abstract
MicroRNAs are important regulators of gene expression. The vast majority of the cells in our body rely on hundreds of these tiny non-coding RNA molecules to precisely adjust their protein repertoire and faithfully accomplish their tasks. Indeed, alterations in the microRNA profile can lead to cellular dysfunction that favours the appearance of several diseases. A specific set of microRNAs plays a crucial role in pancreatic beta cell differentiation and is essential for the fine-tuning of insulin secretion and for compensatory beta cell mass expansion in response to insulin resistance. Recently, several independent studies reported alterations in microRNA levels in the islets of animal models of diabetes and in islets isolated from diabetic patients. Surprisingly, many of the changes in microRNA expression observed in animal models of diabetes were not detected in the islets of diabetic patients and vice versa. These findings are unlikely to merely reflect species differences because microRNAs are highly conserved in mammals. These puzzling results are most probably explained by fundamental differences in the experimental approaches which selectively highlight the microRNAs directly contributing to diabetes development, the microRNAs predisposing individuals to the disease or the microRNAs displaying expression changes subsequent to the development of diabetes. In this review we will highlight the suitability of the different models for addressing each of these questions and propose future strategies that should allow us to obtain a better understanding of the contribution of microRNAs to the development of diabetes mellitus in humans.
Collapse
Affiliation(s)
- Claudiane Guay
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 9, 1005, Lausanne, Switzerland
| | | |
Collapse
|
81
|
Chan KHK, Chacko SA, Song Y, Cho M, Eaton CB, Wu WCH, Liu S. Genetic variations in magnesium-related ion channels may affect diabetes risk among African American and Hispanic American women. J Nutr 2015; 145:418-24. [PMID: 25733456 PMCID: PMC4336527 DOI: 10.3945/jn.114.203489] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Prospective studies consistently link low magnesium intake to higher type 2 diabetes (T2D) risk. OBJECTIVE We examined the association of common genetic variants [single nucleotide polymorphisms (SNPs)] in genes related to magnesium homeostasis with T2D risk and potential interactions with magnesium intake. METHODS Using the Women's Health Initiative-SNP Health Association Resource (WHI-SHARe) study, we identified 17 magnesium-related ion channel genes (583 SNPs) and examined their associations with T2D risk in 7287 African-American (AA; n = 1949 T2D cases) and 3285 Hispanic-American (HA; n = 611 T2D cases) postmenopausal women. We performed both single- and multiple-locus haplotype analyses. RESULTS Among AA women, carriers of each additional copy of SNP rs6584273 in cyclin mediator 1 (CNNM1) had 16% lower T2D risk [OR: 0.84; false discovery rate (FDR)-adjusted P = 0.02]. Among HA women, several variants were significantly associated with T2D risk, including rs10861279 in solute carrier family 41 (anion exchanger), member 2 (SLC41A2) (OR: 0.54; FDR-adjusted P = 0.04), rs7174119 in nonimprinted in Prader-Willi/Angelman syndrome 1 (NIPA1) (OR: 1.27; FDR-adjusted P = 0.04), and 2 SNPs in mitochondrial RNA splicing 2 (MRS2) (rs7738943: OR = 1.55, FDR-adjusted P = 0.01; rs1056285: OR = 1.48, FDR-adjusted P = 0.02). Even with the most conservative Bonferroni adjustment, two 2-SNP-haplotypes in SLC41A2 and MRS2 region were significantly associated with T2D risk (rs12582312-rs10861279: P = 0.0006; rs1056285-rs7738943: P = 0.002). Among women with magnesium intake in the lowest 30% (AA: ≤0.164 g/d; HA: ≤0.185 g/d), 4 SNP signals were strengthened [rs11590362 in claudin 19 (CLDN19), rs823154 in SLC41A1, rs5929706 and rs5930817 in membra; HA: ≥0.313 g/d), rs6584273 in CNNM1 (OR: 0.71; FDR-adjusted P = 0.04) and rs1800467 in potassium inwardly rectifying channel, subfamily J, member 11 (KCNJ11) (OR: 2.50; FDR-adjusted P = 0.01) were significantly associated with T2D risk. CONCLUSIONS Our findings suggest important associations between genetic variations in magnesium-related ion channel genes and T2D risk in AA and HA women that vary by amount of magnesium intake.
Collapse
Affiliation(s)
| | - Sara A Chacko
- Division of General Medicine and Primary Care, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Yiqing Song
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN; Departments of
| | - Michele Cho
- Medicine and,Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA
| | - Charles B Eaton
- Family Medicine,,Epidemiology, and,Center for Primary Care and Prevention, Memorial Hospital of Rhode Island, Pawtucket, RI
| | - Wen-Chih H Wu
- Medicine, Warren Alpert Medical School, Brown University, Providence, RI;,Vascular Research Laboratory, Providence US Department of Veterans Affairs Medical Center, Providence, RI; and
| | - Simin Liu
- Department of Epidemiology, and Departments of Medicine, Warren Alpert Medical School, Brown University, Providence, RI; Medicine and Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA; Division of Endocrinology, Rhode Island Hospital, Providence, RI
| |
Collapse
|
82
|
Exposure to bisphenol A, but not phthalates, increases spontaneous diabetes type 1 development in NOD mice. Toxicol Rep 2015; 2:99-110. [PMID: 28962342 PMCID: PMC5598488 DOI: 10.1016/j.toxrep.2015.02.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/20/2015] [Accepted: 02/22/2015] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune destruction of insulin producing pancreatic beta-cells due to a genetic predisposition and can be triggered by environmental factors. We have previously shown that bisphenol A (BPA) accelerates the spontaneous development of diabetes in non-obese diabetic (NOD) mice. Here, we hypothesized that oral exposure to a mixture of the endocrine disruptors BPA and phthalates, relevant for human exposure, would accelerate diabetes development compared to BPA alone. NOD mice were exposed to BPA (1 mg/l), a mixture of phthalates (DEHP 1 mg/l, DBP 0.2 mg/l, BBP 10 mg/l and DiBP 20 mg/l) or a combination of BPA and the phthalate mixture through drinking water from conception and throughout life. Previous observations that BPA exposure increased the prevalence of diabetes and insulitis and decreased the number of tissue resident macrophages in pancreas were confirmed, and extended by demonstrating that BPA exposure also impaired the phagocytic activity of peritoneal macrophages. None of these effects were observed after phthalate exposure alone. The phthalate exposure in combination with BPA seemed to dampen the BPA effects on macrophage number and function as well as diabetes development, but not insulitis development. Exposure to BPA alone or in combination with phthalates decreased cytokine release (TNFα, IL-6, IL-10, IFNγ, IL-4) from in vitro stimulated splenocytes and lymph node cells, indicating systemic changes in immune function. In conclusion, exposure to BPA, but not to phthalates or mixed exposure to BPA and phthalates, accelerated diabetes development in NOD mice, apparently in part via systemic immune alterations including decreased macrophage function.
Collapse
|
83
|
Farr RJ, Joglekar MV, Hardikar AA. Circulating microRNAs in Diabetes Progression: Discovery, Validation, and Research Translation. EXPERIENTIA SUPPLEMENTUM (2012) 2015; 106:215-244. [PMID: 26608206 DOI: 10.1007/978-3-0348-0955-9_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Diabetes, in all of its forms, is a disease state that demonstrates wide ranging pathological effects throughout the body. Until now, the only method of diagnosing and monitoring the progression of diabetes was through the measurement of blood glucose. Unfortunately, beta cell dysfunction initiates well before the clinical onset of diabetes, and so the development of an effective biomarker signature is of paramount importance to predict and monitor the progression of this disease. MicroRNAs (miRNAs/miRs) are small (18-22 nucleotide) noncoding (nc)RNAs that post-transcriptionally regulate endogenous gene expression by targeted inhibition or degradation of messenger (m)RNA. Recently, miRNAs have shown great promise as biomarkers as some exhibit differential expression in multiple disease states, including type 1 and type 2 diabetes (T1D/T2D). Furthermore, miRNAs are quite stable in circulation, resistant to freeze-thaw and pH-mediated degradation, and are relatively easy to detect using quantitative (q)PCR. Here, we discuss microRNAs that may form a diabetes biomarker signature. To identify these transcripts we outline miRNAs that play a central role in pancreas development and diabetes, as well as previously identified miRNAs with differential expression in individuals with T1D and T2D. Validation and refinement of a miRNA biomarker signature for diabetes would allow identification and intervention of individuals at risk of this disease, as well as stratification and monitoring of patients with established diabetes.
Collapse
Affiliation(s)
- Ryan J Farr
- Diabetes and Islet Biology Group, NHMRC Clinical Trials Centre, Sydney Medical School, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW, 2050, Australia
| | - Mugdha V Joglekar
- Diabetes and Islet Biology Group, NHMRC Clinical Trials Centre, Sydney Medical School, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW, 2050, Australia
| | - Anandwardhan A Hardikar
- Diabetes and Islet Biology Group, NHMRC Clinical Trials Centre, Sydney Medical School, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
84
|
Abstract
The β-cells within the pancreas are responsible for production and secretion of insulin. Insulin is released from pancreatic β-cells in response to increasing blood glucose levels and acts on insulin-sensitive tissues such as skeletal muscle and liver in order to maintain normal glucose homeostasis. Therefore, defects in pancreatic β-cell function lead to hyperglycemia and diabetes mellitus. A new class of molecules called microRNAs has been recently demonstrated to play a crucial role in regulation of pancreatic β-cell function under normal and pathophysiological conditions. miRNAs have been shown to regulate endocrine pancreas development, insulin biosynthesis, insulin exocytosis, and β-cell expansion. Many of the β-cell enriched miRNAs have multiple functions and regulate pancreas development as well as insulin biosynthesis and exocytosis. Furthermore, several of the β-cell specific miRNAs have been shown to accumulate in the circulation before the onset of diabetes and may serve as potential biomarkers for prediabetes. This chapter will focus on miRNAs that are enriched in pancreatic β-cells and play a critical role in modulation of β-cell physiology and may have clinical significance in the treatment of diabetes.
Collapse
|
85
|
Esguerra JLS, Mollet IG, Salunkhe VA, Wendt A, Eliasson L. Regulation of Pancreatic Beta Cell Stimulus-Secretion Coupling by microRNAs. Genes (Basel) 2014; 5:1018-31. [PMID: 25383562 PMCID: PMC4276924 DOI: 10.3390/genes5041018] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/01/2014] [Accepted: 10/21/2014] [Indexed: 12/31/2022] Open
Abstract
Increased blood glucose after a meal is countered by the subsequent increased release of the hypoglycemic hormone insulin from the pancreatic beta cells. The cascade of molecular events encompassing the initial sensing and transport of glucose into the beta cell, culminating with the exocytosis of the insulin large dense core granules (LDCVs) is termed "stimulus-secretion coupling." Impairment in any of the relevant processes leads to insufficient insulin release, which contributes to the development of type 2 diabetes (T2D). The fate of the beta cell, when exposed to environmental triggers of the disease, is determined by the possibility to adapt to the new situation by regulation of gene expression. As established factors of post-transcriptional regulation, microRNAs (miRNAs) are well-recognized mediators of beta cell plasticity and adaptation. Here, we put focus on the importance of comprehending the transcriptional regulation of miRNAs, and how miRNAs are implicated in stimulus-secretion coupling, specifically those influencing the late stages of insulin secretion. We suggest that efficient beta cell adaptation requires an optimal balance between transcriptional regulation of miRNAs themselves, and miRNA-dependent gene regulation. The increased knowledge of the beta cell transcriptional network inclusive of non-coding RNAs such as miRNAs is essential in identifying novel targets for the treatment of T2D.
Collapse
Affiliation(s)
- Jonathan L S Esguerra
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, CRC 91-11, Jan Waldenströms gata 35, 205 02 Malmö, Sweden.
| | - Inês G Mollet
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, CRC 91-11, Jan Waldenströms gata 35, 205 02 Malmö, Sweden.
| | - Vishal A Salunkhe
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, CRC 91-11, Jan Waldenströms gata 35, 205 02 Malmö, Sweden.
| | - Anna Wendt
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, CRC 91-11, Jan Waldenströms gata 35, 205 02 Malmö, Sweden.
| | - Lena Eliasson
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, CRC 91-11, Jan Waldenströms gata 35, 205 02 Malmö, Sweden.
| |
Collapse
|
86
|
Shaer A, Azarpira N, Karimi MH. Differentiation of human induced pluripotent stem cells into insulin-like cell clusters with miR-186 and miR-375 by using chemical transfection. Appl Biochem Biotechnol 2014; 174:242-58. [PMID: 25059983 DOI: 10.1007/s12010-014-1045-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 07/07/2014] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus is characterized by either the inability to produce insulin or insensitivity to insulin secreted by the body. Islet cell replacement is an effective approach for diabetes treatment; however, it is not sufficient for all the diabetic patients. MicroRNAs (miRNAs) are a class of small noncoding RNAs that play an important role in mediating a broad and expanding range of biological activities, such as pancreas development. The present study aimed to develop a protocol to efficiently differentiate human induced pluripotent stem (iPS) cells into islet-like cell clusters (ILCs) in vitro by using miR-186 and miR-375. The human iPS colonies were transfected with hsa-miR-186 and hsa-miR-375 by using siPORT™ NeoFX™ Transfection Agent, and the differentiation was compared to controls. Total RNA was extracted 24 and 48 h after transfection. The gene expressions of insulin, NGN3, GLUT2, PAX4, PAX6, KIR6.2, NKX6.1, PDX1, Glucagon, and OCT4 were then evaluated through real-time qPCR. On the third day, the potency of the clusters was assessed in response to high glucose levels. Dithizone (DTZ) was used to identify the existence of the β-cells. Besides, the presence of insulin and NGN3 proteins was investigated by immunocytochemistry. Morphological changes were observed on the first day after the chemical transfection, and cell clusters were formed on the third day. The expression of pancreatic specific transcription factors was increased on the first day and significantly increased on the second day. The ILCs were positive for insulin and NGN3 proteins in the immunocytochemistry. Besides, the clusters were stained with DTZ and secreted insulin in glucose challenge test. Overexpression of miR-186 and miR-375 can be an alternative strategy for producing ILCs from the iPS cells in a short time. This work provides a new approach by using patient-specific iPSCs for β-cell replacement therapy in diabetic patients.
Collapse
Affiliation(s)
- Anahita Shaer
- Department of Biology, Science and Research Branch, Islamic Azad University, Fars, Iran
| | | | | |
Collapse
|
87
|
Price NL, Ramírez CM, Fernández-Hernando C. Relevance of microRNA in metabolic diseases. Crit Rev Clin Lab Sci 2014; 51:305-20. [PMID: 25034902 DOI: 10.3109/10408363.2014.937522] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Metabolic syndrome is a complex metabolic condition caused by abnormal adipose deposition and function, dyslipidemia and hyperglycemia, which affects >47 million American adults and ∼1 million children. Individuals with the metabolic syndrome have essentially twice the risk for developing cardiovascular disease (CVD) and Type 2 diabetes mellitus (T2D), compared to those without the syndrome. In the search for improved and novel therapeutic strategies, microRNAs (miRNA) have been shown to be interesting targets due to their regulatory role on gene networks controlling different crucial aspects of metabolism, including lipid and glucose homeostasis. More recently, the discovery of circulating miRNAs suggest that miRNAs may be involved in facilitating metabolic crosstalk between organs as well as serving as novel biomarkers of diseases, including T2D and atherosclerosis. These findings highlight the importance of miRNAs for regulating pathways that underlie metabolic diseases, and their potential as therapeutic targets for the development of novel treatments.
Collapse
|
88
|
Kaspi H, Pasvolsky R, Hornstein E. Could microRNAs contribute to the maintenance of β cell identity? Trends Endocrinol Metab 2014; 25:285-92. [PMID: 24656914 DOI: 10.1016/j.tem.2014.01.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 01/21/2014] [Accepted: 01/29/2014] [Indexed: 12/22/2022]
Abstract
Normal physiology depends on defined functional output of differentiated cells. However, differentiated cells are often surprisingly fragile. As an example, phenotypic collapse and dedifferentiation of β cells were recently discovered in the pathogenesis of type 2 diabetes (T2D). These discoveries necessitate the investigation of mechanisms that function to maintain robust cell type identity. microRNAs (miRNAs), which are small non-coding RNAs, are known to impart robustness to development. miRNAs are interlaced within networks, that include also transcriptional and epigenetic regulators, for continuous control of lineage-specific gene expression. In this Opinion article, we provide a framework for conceptualizing how miRNAs might participate in adult β cell identity and suggest that miRNAs may function as important genetic components in metabolic disorders, including diabetes.
Collapse
Affiliation(s)
- Haggai Kaspi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ronit Pasvolsky
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Eran Hornstein
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
89
|
Eliasson L, Esguerra JLS. Role of non-coding RNAs in pancreatic beta-cell development and physiology. Acta Physiol (Oxf) 2014; 211:273-84. [PMID: 24666639 DOI: 10.1111/apha.12285] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 02/25/2014] [Accepted: 03/17/2014] [Indexed: 12/15/2022]
Abstract
The progression of diabetes is accompanied by increasing demand to the beta-cells to produce and secrete more insulin, requiring complex beta-cell adaptations. Functionally active and ubiquitous non-coding RNAs (ncRNAs) have the capacity to take part in such adaptations as they have been shown to be key regulatory molecules in various biological processes. In the pancreatic islets, the function of ncRNAs and their contribution to disease development is beginning to be understood. Here, we review the different classes of ncRNAs, such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), and their potential contribution to insulin secretion. A special focus will be on miRNAs and their regulatory function in beta-cell physiology and insulin exocytosis. As important players in gene regulation, ncRNAs have huge potential in opening innovative therapeutic avenues against diabetes and associated complications.
Collapse
Affiliation(s)
- L. Eliasson
- Department of Clinical Sciences-Malmö; Islet Cell Exocytosis; Lund University Diabetes Centre; Lund University; Malmö Sweden
| | - J. L. S. Esguerra
- Department of Clinical Sciences-Malmö; Islet Cell Exocytosis; Lund University Diabetes Centre; Lund University; Malmö Sweden
| |
Collapse
|
90
|
Huang JT, Wang J, Srivastava V, Sen S, Liu SM. MicroRNA Machinery Genes as Novel Biomarkers for Cancer. Front Oncol 2014; 4:113. [PMID: 24904827 PMCID: PMC4032885 DOI: 10.3389/fonc.2014.00113] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/01/2014] [Indexed: 12/25/2022] Open
Abstract
MicroRNAs (miRNAs) directly and indirectly affect tumorigenesis. To be able to perform their myriad roles, miRNA machinery genes, such as Drosha, DGCR8, Dicer1, XPO5, TRBP, and AGO2, must generate precise miRNAs. These genes have specific expression patterns, protein-binding partners, and biochemical capabilities in different cancers. Our preliminary analysis of data from The Cancer Genome Atlas consortium on multiple types of cancer revealed significant alterations in these miRNA machinery genes. Here, we review their biological structures and functions with an eye toward understanding how they could serve as cancer biomarkers.
Collapse
Affiliation(s)
- Jing-Tao Huang
- Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University , Wuhan , China
| | - Jin Wang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Vibhuti Srivastava
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Subrata Sen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Song-Mei Liu
- Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University , Wuhan , China
| |
Collapse
|
91
|
Plaisance V, Waeber G, Regazzi R, Abderrahmani A. Role of microRNAs in islet beta-cell compensation and failure during diabetes. J Diabetes Res 2014; 2014:618652. [PMID: 24734255 PMCID: PMC3964735 DOI: 10.1155/2014/618652] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 01/24/2014] [Indexed: 12/12/2022] Open
Abstract
Pancreatic beta-cell function and mass are markedly adaptive to compensate for the changes in insulin requirement observed during several situations such as pregnancy, obesity, glucocorticoids excess, or administration. This requires a beta-cell compensation which is achieved through a gain of beta-cell mass and function. Elucidating the physiological mechanisms that promote functional beta-cell mass expansion and that protect cells against death, is a key therapeutic target for diabetes. In this respect, several recent studies have emphasized the instrumental role of microRNAs in the control of beta-cell function. MicroRNAs are negative regulators of gene expression, and are pivotal for the control of beta-cell proliferation, function, and survival. On the one hand, changes in specific microRNA levels have been associated with beta-cell compensation and are triggered by hormones or bioactive peptides that promote beta-cell survival and function. Conversely, modifications in the expression of other specific microRNAs contribute to beta-cell dysfunction and death elicited by diabetogenic factors including, cytokines, chronic hyperlipidemia, hyperglycemia, and oxidized LDL. This review underlines the importance of targeting the microRNA network for future innovative therapies aiming at preventing the beta-cell decline in diabetes.
Collapse
Affiliation(s)
- Valérie Plaisance
- Lille 2 University, European Genomic Institute for Diabetes (EGID), FR 3508, UMR-8199 Lille, France
| | - Gérard Waeber
- Service of Internal Medicine, Hospital-University of Lausanne (CHUV), 1011 Lausanne, Switzerland
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Amar Abderrahmani
- Lille 2 University, European Genomic Institute for Diabetes (EGID), FR 3508, UMR-8199 Lille, France
- *Amar Abderrahmani:
| |
Collapse
|
92
|
Reinbothe TM, Safi F, Axelsson AS, Mollet IG, Rosengren AH. Optogenetic control of insulin secretion in intact pancreatic islets with β-cell-specific expression of Channelrhodopsin-2. Islets 2014; 6:e28095. [PMID: 25483880 PMCID: PMC4593566 DOI: 10.4161/isl.28095] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Insulin is secreted from the pancreatic β-cells in response to elevated glucose. In intact islets the capacity for insulin release is determined by a complex interplay between different cell types. This has made it difficult to specifically assess the role of β-cell defects to the insulin secretory impairment in type 2 diabetes. Here we describe a new approach, based on optogenetics, that enables specific investigation of β-cells in intact islets. We used transgenic mice expressing the light-sensitive cation channel Channelrhodopsin-2 (ChR2) under control of the insulin promoter. Glucose tolerance in vivo was assessed using intraperitoneal glucose tolerance tests, and glucose-induced insulin release was measured from static batch incubations. ChR2 localization was determined by fluorescence confocal microscopy. The effect of ChR2 stimulation with blue LED light was assessed using Ca(2+) imaging and static islet incubations. Light stimulation of islets from transgenic ChR2 mice triggered prompt increases in intracellular Ca(2+). Moreover, light stimulation enhanced insulin secretion in batch-incubated islets at low and intermediate but not at high glucose concentrations. Glucagon release was not affected. Beta-cells from mice rendered diabetic on a high-fat diet exhibited a 3.5-fold increase in light-induced Ca(2+) influx compared with mice on a control diet. Furthermore, light enhanced insulin release also at high glucose in these mice, suggesting that high-fat feeding leads to a compensatory potentiation of the Ca(2+) response in β-cells. The results demonstrate the usefulness and versatility of optogenetics for studying mechanisms of perturbed hormone secretion in diabetes with high time-resolution and cell-specificity.
Collapse
|
93
|
Dere E, Anderson LM, Hwang K, Boekelheide K. Biomarkers of chemotherapy-induced testicular damage. Fertil Steril 2013; 100:1192-202. [PMID: 24182554 DOI: 10.1016/j.fertnstert.2013.09.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/27/2013] [Accepted: 09/12/2013] [Indexed: 12/16/2022]
Abstract
Increasing numbers of men are having or wanting children after chemotherapy treatment. This can be attributed to improvements in cancer therapies that increase survival. However, a side effect of most chemotherapy drugs is disruption of spermatogenesis and a drastic reduction in sperm count and quality. Although many men eventually recover reproductive function, as indicated by normal semen analyses, there is no clinical test that can assess sperm quality at a high level of sensitivity. Sperm fluorescent in situ hybridization (i.e., FISH) and several different tests for deoxyribonucleic acid (DNA) fragmentation have been used infrequently in clinical assessment. Animal models of chemotherapy-induced testicular damage are currently being used to identify potential molecular biomarkers that may be translatable to humans-these include sperm messenger RNAs, microRNAs, histone modifications, and DNA methylation patterns. Changes in these molecular measurements are quantitative and sensitive, potentially making them important clinical biomarkers of testicular function after chemotherapy treatment.
Collapse
Affiliation(s)
- Edward Dere
- Division of Urology, Rhode Island Hospital, Providence, Rhode Island; Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | | | | | | |
Collapse
|
94
|
Abstract
Epigenetic mechanisms, including DNA methylation, histone modifications, and noncoding RNA expression, contribute to regulate islet cell development and function. Indeed, epigenetic mechanisms were recently shown to be involved in the control of endocrine cell fate decision, islet differentiation, β-cell identity, proliferation, and mature function. Epigenetic mechanisms can also contribute to the pathogenesis of complex diseases. Emerging knowledge regarding epigenetic mechanisms suggest that they may be involved in β-cell dysfunction and pathogenesis of diabetes. Epigenetic mechanisms could predispose to the diabetic phenotype such as decline of β-cell proliferation ability and β-cell failure, and account for complications associated with diabetes. Better understanding of epigenetic landscapes of islet differentiation and function may be useful to improve β-cell differentiation protocols and discover novel therapeutic targets for prevention and treatment of diabetes.
Collapse
Affiliation(s)
- Cecile Haumaitre
- CNRS UMR 7622, Université Pierre et Marie Curie, INSERM U969, 9 quai Saint Bernard, 75005, Paris, France,
| |
Collapse
|
95
|
Dere E, Anderson LM, Coulson M, McIntyre BS, Boekelheide K, Chapin RE. SOT Symposium Highlight: Translatable Indicators of Testicular Toxicity: Inhibin B, MicroRNAs, and Sperm Signatures. Toxicol Sci 2013; 136:265-73. [PMID: 24052563 DOI: 10.1093/toxsci/kft207] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Testicular toxicity is an important safety endpoint in drug development and risk assessment, but reliable and translatable biomarkers for predicting injury have eluded researchers. However, this area shows great potential for improvement, with several avenues currently being pursued. This was the topic of a symposium session during the 2013 Society of Toxicology Annual Meeting in San Antonio, TX, entitled "Translatable Indicators of Testicular Toxicity: Inhibin B, MicroRNAs, and Sperm Signatures." This symposium brought together stakeholders from academia, government, and industry to present the limitations and drawbacks of currently used indicators of injury and discussed the ongoing efforts in developing more predictive biomarkers of injury. The presentations highlighted the early challenges of using circulating inhibin B and microRNA levels, and sperm messenger RNA transcript abundance and DNA methylation profiles, as novel biomarkers of testicular toxicity.
Collapse
Affiliation(s)
- Edward Dere
- * Division of Urology, Rhode Island Hospital, Providence, Rhode Island 02903
| | | | | | | | | | | |
Collapse
|
96
|
MicroRNAs: new insights into chronic childhood diseases. BIOMED RESEARCH INTERNATIONAL 2013; 2013:291826. [PMID: 23878802 PMCID: PMC3710618 DOI: 10.1155/2013/291826] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 06/07/2013] [Indexed: 02/06/2023]
Abstract
Chronic diseases are the major cause of morbidity and mortality worldwide and have shown increasing incidence rates among children in the last decades. Chronic illnesses in the pediatric population, even if well managed, affect social, psychological, and physical development and often limit education and active participation and increase the risk for health complications. The significant pediatric morbidity and mortality rates caused by chronic illnesses call for serious efforts toward better understanding of the pathogenesis of these disorders. Recent studies have shown the involvement of microRNAs (miRNAs) in various aspects of major pediatric chronic non-neoplastic diseases. This review focuses on the role of miRNAs in four major pediatric chronic diseases including bronchial asthma, diabetes mellitus, epilepsy and cystic fibrosis. We intend to emphasize the importance of miRNA-based research in combating these major disorders, as we believe this approach will result in novel therapies to aid securing normal development and to prevent disabilities in the pediatric population.
Collapse
|
97
|
Abstract
MicroRNAs (miRNAs) are important regulators of gene expression programs in the pancreas; however, little is known about the role of miRNA pathways during endocrine cell specification and maturation during neonatal life. In this study, we deleted Dicer1, an essential RNase for active miRNAs biogenesis, specifically from NGN3+ endocrine progenitor cells. We found that deletion of Dicer1 in endocrine progenitors did not affect the specification of hormone-expressing endocrine cells. However, the islets in the mutant mice in the neonatal period exhibited morphological defects in organization and loss of hormone expression, and the mutant mice subsequently developed diabetes. Dicer1-deficient β-cells lost insulin expression while maintaining the expression of β-cell transcription factors such as Pdx1 and Nkx6.1 early in the postnatal period. Surprisingly, transcriptional profiling showed that that the Dicer1-deficient endocrine cells expressed neuronal genes before the onset of diabetes. The derepression of neuronal genes was associated with a loss in binding of the neuronal transcriptional repressor RE-1-silencing transcription factor to its targets in Dicer1-deficient β-cells. These studies suggest that miRNAs play a critical role in suppressing neuronal genes during the maturation of endocrine cells.
Collapse
Affiliation(s)
- Murtaza S. Kanji
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Molecular Biology Interdepartmental Ph.D. Program (MBIDP), University of California, Los Angeles, Los Angeles, California
| | - Martin G. Martin
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Anil Bhushan
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Molecular Biology Interdepartmental Ph.D. Program (MBIDP), University of California, Los Angeles, Los Angeles, California
- Molecular, Cellular and Developmental Biology, University of California, Los Angeles, Los Angeles, California
- Corresponding author: Anil Bhushan,
| |
Collapse
|
98
|
Klein D, Misawa R, Bravo-Egana V, Vargas N, Rosero S, Piroso J, Ichii H, Umland O, Zhijie J, Tsinoremas N, Ricordi C, Inverardi L, Domínguez-Bendala J, Pastori RL. MicroRNA expression in alpha and beta cells of human pancreatic islets. PLoS One 2013; 8:e55064. [PMID: 23383059 PMCID: PMC3558471 DOI: 10.1371/journal.pone.0055064] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 12/22/2012] [Indexed: 12/26/2022] Open
Abstract
microRNAs (miRNAs) play an important role in pancreatic development and adult β-cell physiology. Our hypothesis is based on the assumption that each islet cell type has a specific pattern of miRNA expression. We sought to determine the profile of miRNA expression in α-and β-cells, the main components of pancreatic islets, because this analysis may lead to a better understanding of islet gene regulatory pathways. Highly enriched (>98%) subsets of human α-and β-cells were obtained by flow cytometric sorting after intracellular staining with c-peptide and glucagon antibody. The method of sorting based on intracellular staining is possible because miRNAs are stable after fixation. MiRNA expression levels were determined by quantitative high throughput PCR-based miRNA array platform screening. Most of the miRNAs were preferentially expressed in β-cells. From the total of 667 miRNAs screened, the Significant Analysis of Microarray identified 141 miRNAs, of which only 7 were expressed more in α-cells (α-miRNAs) and 134 were expressed more in β-cells (β-miRNAs). Bioinformatic analysis identified potential targets of β-miRNAs analyzing the Beta Cell Gene Atlas, described in the T1Dbase, the web platform, supporting the type 1 diabetes (T1D) community. cMaf, a transcription factor regulating glucagon expression expressed selectively in α-cells (TFα) is targeted by β-miRNAs; miR-200c, miR-125b and miR-182. Min6 cells treated with inhibitors of these miRNAs show an increased expression of cMaf RNA. Conversely, over expression of miR-200c, miR-125b or miR-182 in the mouse alpha cell line αTC6 decreases the level of cMAF mRNA and protein. MiR-200c also inhibits the expression of Zfpm2, a TFα that inhibits the PI3K signaling pathway, at both RNA and protein levels.In conclusion, we identified miRNAs differentially expressed in pancreatic α- and β-cells and their potential transcription factor targets that could add new insights into different aspects of islet biology and pathophysiology.
Collapse
Affiliation(s)
- Dagmar Klein
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Ryosuke Misawa
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Valia Bravo-Egana
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Nancy Vargas
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Samuel Rosero
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Julieta Piroso
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Hirohito Ichii
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Oliver Umland
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Jiang Zhijie
- Center for Computational Science, University of Miami, Miami, Florida, United States of America
| | - Nicholas Tsinoremas
- Center for Computational Science, University of Miami, Miami, Florida, United States of America
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
- Department of Surgical Sciences, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Luca Inverardi
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
- Department of Surgical Sciences, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Ricardo L. Pastori
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
99
|
MicroRNA expression profiling of human islets from individuals with and without type 2 diabetes: promises and pitfalls. Biochem Soc Trans 2012; 40:800-3. [PMID: 22817737 DOI: 10.1042/bst20120049] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Recent studies in mouse, involving the β-cell-specific deletion of Dicer1, have highlighted the crucial role of miRNAs (microRNAs) in regulating insulin secretion and consequently Type 2 diabetes. Identifying the individual miRNAs involved in human islet dysfunction may be of diagnostic and therapeutic interest. miRNA expression profiling of human islets isolated from donors with and without Type 2 diabetes may represent one of the first steps in the discovery of these specific miRNAs. The present review discusses some of the potential pitfalls and promises of such an approach.
Collapse
|
100
|
Kumar M, Nath S, Prasad HK, Sharma GD, Li Y. MicroRNAs: a new ray of hope for diabetes mellitus. Protein Cell 2012; 3:726-38. [PMID: 23055040 DOI: 10.1007/s13238-012-2055-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 07/01/2012] [Indexed: 12/28/2022] Open
Abstract
Diabetes mellitus has become one of the most common chronic diseases, thereby posing a major challenge to global health. Characterized by high levels of blood glucose (hyperglycemia), diabetes usually results from a loss of insulin-producing β-cells in the pancreas, leading to a deficiency of insulin (type 1 diabetes), or loss of insulin sensitivity (type 2 diabetes). Both types of diabetes have serious secondary complications, such as microvascular abnormalities, cardiovascular dysfunction, and kidney failure. Various complex factors, such as genetic and environmental factors, are associated with the pathophysiology of diabetes. Over the past two decades, the role of small, single-stranded noncoding microRNAs in various metabolic disorders, especially diabetes mellitus and its complications, has gained widespread attention in the scientific community. Discovered first as an endogenous regulator of development in the nematode Caenorhabditis elegans, these small RNAs post-transcriptionally suppress mRNA target expression. In this review, we discuss the potential roles of different microRNAs in diabetes and diabetes-related complications.
Collapse
Affiliation(s)
- Munish Kumar
- Department of Biotechnology, Assam University, Silchar, India.
| | | | | | | | | |
Collapse
|