1
|
Carr ER, Higgins PB, McClenaghan NH, Flatt PR, McCloskey AG. MicroRNA regulation of islet and enteroendocrine peptides: Physiology and therapeutic implications for type 2 diabetes. Peptides 2024; 176:171196. [PMID: 38492669 DOI: 10.1016/j.peptides.2024.171196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/05/2024] [Accepted: 03/14/2024] [Indexed: 03/18/2024]
Abstract
The pathogenesis of type 2 diabetes (T2D) is associated with dysregulation of glucoregulatory hormones, including both islet and enteroendocrine peptides. Microribonucleic acids (miRNAs) are short noncoding RNA sequences which post transcriptionally inhibit protein synthesis by binding to complementary messenger RNA (mRNA). Essential for normal cell activities, including proliferation and apoptosis, dysregulation of these noncoding RNA molecules have been linked to several diseases, including diabetes, where alterations in miRNA expression within pancreatic islets have been observed. This may occur as a compensatory mechanism to maintain beta-cell mass/function (e.g., downregulation of miR-7), or conversely, lead to further beta-cell demise and disease progression (e.g., upregulation of miR-187). Thus, targeting miRNAs has potential for novel diagnostic and therapeutic applications in T2D. This is reinforced by the success seen to date with miRNA-based therapeutics for other conditions currently in clinical trials. In this review, differential expression of miRNAs in human islets associated with T2D will be discussed along with further consideration of their effects on the production and secretion of islet and incretin hormones. This analysis further unravels the therapeutic potential of miRNAs and offers insights into novel strategies for T2D management.
Collapse
Affiliation(s)
- E R Carr
- Department of Life and Physical Sciences, Atlantic Technology University, Donegal, Ireland; Department of Life Sciences, Atlantic Technological University, Sligo, Ireland
| | - P B Higgins
- Department of Life and Physical Sciences, Atlantic Technology University, Donegal, Ireland
| | - N H McClenaghan
- Department of Life Sciences, Atlantic Technological University, Sligo, Ireland
| | - P R Flatt
- School of Biomedical Sciences, Ulster University, Coleraine, UK
| | - A G McCloskey
- Department of Life and Physical Sciences, Atlantic Technology University, Donegal, Ireland.
| |
Collapse
|
2
|
Wu W, Ren J, Wang J, Wang J, Yu D, Zhang Y, Zeng F, Huang B. Metalloestrogens exposure and risk of gestational diabetes mellitus: Evidence emerging from the systematic review and meta-analysis. ENVIRONMENTAL RESEARCH 2024; 248:118321. [PMID: 38307186 DOI: 10.1016/j.envres.2024.118321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/04/2024] [Accepted: 01/25/2024] [Indexed: 02/04/2024]
Abstract
BACKGROUND Metalloestrogens are metals and metalloid elements with estrogenic activity found everywhere. Their impact on human health is becoming more apparent as human activities increase. OBJECTIVE Our aim is to conduct a comprehensive systematic review and meta-analysis of observational studies exploring the correlation between metalloestrogens (specifically As, Sb, Cr, Cd, Cu, Se, Hg) and Gestational Diabetes Mellitus (GDM). METHODS PubMed, Web of Science, and Embase were searched to examine the link between metalloestrogens (As, Sb, Cr, Cd, Cu, Se, and Hg) and GDM until December 2023. Risk estimates were derived using random effects models. Subgroup analyses were conducted based on study countries, exposure sample, exposure assessment method, and detection methods. Sensitivity analyses and adjustments for publication bias were carried out to assess the strength of the findings. RESULTS Out of the 389 articles identified initially, 350 met our criteria and 33 were included in the meta-analysis, involving 141,175 subjects (9450 cases, 131,725 controls). Arsenic, antimony, and copper exposure exhibited a potential increase in GDM risk to some extent (As: OR = 1.28, 95 % CI [1.08, 1.52]; Sb: OR = 1.73, 95 % CI [1.13, 2.65]; Cu: OR = 1.29, 95 % CI [1.02, 1.63]), although there is a high degree of heterogeneity (As: Q = 52.93, p < 0.05, I2 = 64.1 %; Sb: Q = 31.40, p < 0.05, I2 = 80.9 %; Cu: Q = 21.14, p < 0.05, I2 = 71.6 %). Conversely, selenium, cadmium, chromium, and mercury exposure did not exhibit any association with the risk of GDM in our study. DISCUSSION Our research indicates that the existence of harmful metalloestrogens in the surroundings has a notable effect on the likelihood of GDM. Hence, we stress the significance of environmental elements in the development of GDM and the pressing need for relevant policies and measures.
Collapse
Affiliation(s)
- Wanxin Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Junjie Ren
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Juan Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Jiamei Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Deshui Yu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yan Zhang
- School of Biology and Food Engineering, Hefei Normal University, Hefei, 230092, Anhui, China.
| | - Fa Zeng
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 518109, Guangdong, China.
| | - Binbin Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
3
|
Heris HV, Zahraei Z. miRNAs: Regulators of immune system in diabetes. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2022; 790:108442. [PMID: 36089265 DOI: 10.1016/j.mrrev.2022.108442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 01/01/2023]
Abstract
Diabetes, one of the most common multifactorial metabolic disorders, is a jeopardizing cause of human health worldwide. MicroRNAs (miRNAs) are a group of small non-coding RNAs that have been contributed to the regulation of gene expression through post-transcriptional mechanisms. The potential role of miRNAs has been studied in the most of biological processes and mechanisms underlying the progression of variety diseases including diabetes. In this review, we focus on the role of miRNAs in regulating pivotal molecular and cellular mechanisms associated with immune system that progress diabetic disorders.
Collapse
Affiliation(s)
- Helaleh Vaezi Heris
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Zohreh Zahraei
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Iran.
| |
Collapse
|
4
|
Jeje SO, Adenawoola M, Abosede C. Gestational Nutrition as a Predisposing Factor to Obesity Onset in Offspring: Role for Involvement of Epigenetic Mechanism. Niger J Physiol Sci 2022; 37:1-7. [PMID: 35947841 DOI: 10.54548/njps.v37i1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 04/12/2022] [Indexed: 06/15/2023]
Abstract
Maternal lifestyle has been implicated as a predisposing factor in the development of metabolic disorders in adulthood. This lifestyle includes the immediate environment, physical activity and nutrition. Maternal nutrition has direct influence on the developmental programming through biochemical alterations and can lead to modifications in the fetal genome through epigenetic mechanisms. Imbalance in basic micro or macro nutrients due to famine or food deficiency during delicate gestational periods can lead to onset of metabolic syndrome including obesity. A major example is the Dutch famine which led to a serious metabolic disorder in adulthood of affected infants. Notably due to gene variants, individualized responses to nutritional deficiencies are unconventional, therefore intensifying the need to study nutritional genomics during fetal programming. Epigenetic mechanisms can cause hereditary changes without changing the DNA sequence; the major mechanisms include small non-coding RNAs, histone modifications and most stable of all is DNA methylation. The significance association between obesity and DNA methylation is through regulation of genes implicated in lipid and glucose metabolism either directly or indirectly by hypomethylation or hypermethylation. Examples include CPT1A, APOA2, ADRB3 and POMC. Any maternal exposure to malnutrition or overnutrition that can affect genes regulating major metabolic pathways in the fetus, will eventually cause underlying changes that can predispose or cause the onset of metabolic disorder in adulthood. In this review, we examined the interaction between nutrition during gestation and epigenetic programming of metabolic syndrome.
Collapse
|
5
|
Deng S, Yang L, Ma K, Bian W. Astragalus polysaccharide improve the proliferation and insulin secretion of mouse pancreatic β cells induced by high glucose and palmitic acid partially through promoting miR-136-5p and miR-149-5p expression. Bioengineered 2021; 12:9872-9884. [PMID: 34699323 PMCID: PMC8810136 DOI: 10.1080/21655979.2021.1996314] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The current drugs for the treatment of type 2 diabetes mellitus (T2DM) can cause side effects after long-time use. Hence, the novel drugs were urgent need to developed for T2DM patients. In this study, the effect of astragalus polysaccharide on dysfunctional insulin cells was investigated to clarify whether astragalus polysaccharide could be a novel drug for T2DM treatment. MIN6 cells (mouse pancreatic β-cell line) were treated with high glucose (HG)+ palmitic acid (PA) and then treated with astragalus polysaccharide. The proliferation, apoptosis, and insulin secretion were measured using CCK8, flow cytometry, and ELISA, respectively. Pancreatic and duodenal homeobox 1 (PDX1), miR-136-5p, and miR-149-5p expression levels were measured by RT-qPCR. The combination of EF-hand domain family member 2 (EFHD2) and miR-136-5p or miR-149-5p was analyzed by luciferase reporter assay. EFHD2 protein level was measured by western blot. We found that HG+PA treatment reduced MIN6 cell viability, insulin secretion, and PDX1 expression and promoted MIN6 cell apoptosis. Astragalus polysaccharide treatment reversed the effect of HG+PA on MIN6 cells. Additionally, astragalus polysaccharide treatment promoted miR-136-5p and miR-149-5p expression. Silencing of miR-136-5p and miR-149-5p expression partially reversed the therapeutic effects of astragalus polysaccharide. Furthermore, EFHD2 was the target of miR-136-5p and miR-149-5p. Meanwhile, astragalus polysaccharide treatment inhibited EFHD2 protein level in HG+PA treated MIN6 cell. Finally, EFHD2 overexpression partially reversed the therapeutic effects of astragalus polysaccharide. In conclusion, astragalus polysaccharide treatment improved proliferation and insulin secretion in HG+PA-treated MIN6 cells partially by promoting miR-136-5p and miR-149-5p expression to inhibit EFHD2 expression.
Collapse
Affiliation(s)
- Shifang Deng
- Department of Traditional Chinese Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Lei Yang
- Department of Geriatrics in Luohu Hospital of Traditional Chinese Medicine, Shenzhen Hospital of Shanghai University of traditional Chinese Medicine, Shenzhen, China
| | - Ke Ma
- Department of Traditional Chinese Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Wei Bian
- Department of Traditional Chinese Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| |
Collapse
|
6
|
Lin YCD, Huang HY, Shrestha S, Chou CH, Chen YH, Chen CR, Hong HC, Li J, Chang YA, Chiew MY, Huang YR, Tu SJ, Sun TH, Weng SL, Tseng CP, Huang HD. Multi-omics profiling reveals microRNA-mediated insulin signaling networks. BMC Bioinformatics 2020; 21:389. [PMID: 32938376 PMCID: PMC7496206 DOI: 10.1186/s12859-020-03678-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background MicroRNAs (miRNAs) play a key role in mediating the action of insulin on cell growth and the development of diabetes. However, few studies have been conducted to provide a comprehensive overview of the miRNA-mediated signaling network in response to glucose in pancreatic beta cells. In our study, we established a computational framework integrating multi-omics profiles analyses, including RNA sequencing (RNA-seq) and small RNA sequencing (sRNA-seq) data analysis, inverse expression pattern analysis, public data integration, and miRNA targets prediction to illustrate the miRNA-mediated regulatory network at different glucose concentrations in INS-1 pancreatic beta cells (INS-1), which display important characteristics of the pancreatic beta cells. Results We applied our computational framework to the expression profiles of miRNA/mRNA of INS-1, at different glucose concentrations. A total of 1437 differentially expressed genes (DEGs) and 153 differentially expressed miRNAs (DEmiRs) were identified from multi-omics profiles. In particular, 121 DEmiRs putatively regulated a total of 237 DEGs involved in glucose metabolism, fatty acid oxidation, ion channels, exocytosis, homeostasis, and insulin gene regulation. Moreover, Argonaute 2 immunoprecipitation sequencing, qRT-PCR, and luciferase assay identified Crem, Fn1, and Stc1 are direct targets of miR-146b and elucidated that miR-146b acted as a potential regulator and promising target to understand the insulin signaling network. Conclusions In this study, the integration of experimentally verified data with system biology framework extracts the miRNA network for exploring potential insulin-associated miRNA and their target genes. The findings offer a potentially significant effect on the understanding of miRNA-mediated insulin signaling network in the development and progression of pancreatic diabetes.
Collapse
Affiliation(s)
- Yang-Chi-Dung Lin
- School of Life and Health Sciences, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China.,Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China
| | - Hsi-Yuan Huang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China.,Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China
| | - Sirjana Shrestha
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan.,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Chih-Hung Chou
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan.,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Yen-Hua Chen
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, 10021, USA
| | - Chi-Ru Chen
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Hsiao-Chin Hong
- School of Life and Health Sciences, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China.,Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China
| | - Jing Li
- School of Life and Health Sciences, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China.,Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China
| | - Yi-An Chang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Men-Yee Chiew
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Ya-Rong Huang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Siang-Jyun Tu
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Ting-Hsuan Sun
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Shun-Long Weng
- Department of Obstetrics and Gynecology, Hsinchu Mackay Memorial Hospital, Hsinchu, 300, Taiwan
| | - Ching-Ping Tseng
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan.
| | - Hsien-Da Huang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China. .,Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China. .,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan.
| |
Collapse
|
7
|
Cheng L, Zhou M, Zhang D, Chen B. Association of miR-146a polymorphism rs2910164 and type 2 diabetes risk: a meta-analysis. J Int Med Res 2020; 48:300060520931313. [PMID: 32812451 PMCID: PMC7441291 DOI: 10.1177/0300060520931313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Circulating miR-146a is aberrantly expressed in patients with type 2 diabetes (T2D), probably resulting from gene polymorphisms. However, the role of polymorphism rs2910164 in T2D pathogenesis remains controversial. Thus, we designed a meta-analysis to investigate the association between rs2910164 and T2D. METHODS PubMed and Embase were searched for eligible papers in English published through September 2, 2019. Random or fixed effect models were used to determine risk estimates according to heterogeneities. RESULTS Four studies, involving 2,069 patients and 1,950 controls, were included. Odds ratios (ORs) and 95% confidence intervals (95% CIs) were used to pool the effect size. The pooled ORs and 95% CIs were 1.501 (0.887-2.541), 1.102 (0.931-1.304), 1.276 (0.900-1.811), 1.204 (0.878-1.652), 1.238 (0.880-1.740), and 1.350 (0.904-2.016) under the homozygote, heterozygote (CG vs. GG and CC vs. CG), dominant, allele, and recessive models, respectively. Heterogeneity was detected in most genetic models, with subgroup analyses performed by ethnicity, genotyping method, and disease duration. The co-dominant model was determined to be the most appropriate genetic model. CONCLUSIONS Our findings suggested that polymorphism rs2910164 is not correlated with T2D susceptibility. However, the results should be interpreted with caution because of confounding factors.
Collapse
Affiliation(s)
- Liqing Cheng
- Department of Endocrinology and Metabolism, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Zhou
- Department of Urology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dongmei Zhang
- Department of Dermatology, Chongqing MyLike Plastic Surgery Hospital, Chongqing, China
| | - Bing Chen
- Department of Endocrinology and Metabolism, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
8
|
Guay C, Jacovetti C, Bayazit MB, Brozzi F, Rodriguez-Trejo A, Wu K, Regazzi R. Roles of Noncoding RNAs in Islet Biology. Compr Physiol 2020; 10:893-932. [PMID: 32941685 DOI: 10.1002/cphy.c190032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The discovery that most mammalian genome sequences are transcribed to ribonucleic acids (RNA) has revolutionized our understanding of the mechanisms governing key cellular processes and of the causes of human diseases, including diabetes mellitus. Pancreatic islet cells were found to contain thousands of noncoding RNAs (ncRNAs), including micro-RNAs (miRNAs), PIWI-associated RNAs, small nucleolar RNAs, tRNA-derived fragments, long non-coding RNAs, and circular RNAs. While the involvement of miRNAs in islet function and in the etiology of diabetes is now well documented, there is emerging evidence indicating that other classes of ncRNAs are also participating in different aspects of islet physiology. The aim of this article will be to provide a comprehensive and updated view of the studies carried out in human samples and rodent models over the past 15 years on the role of ncRNAs in the control of α- and β-cell development and function and to highlight the recent discoveries in the field. We not only describe the role of ncRNAs in the control of insulin and glucagon secretion but also address the contribution of these regulatory molecules in the proliferation and survival of islet cells under physiological and pathological conditions. It is now well established that most cells release part of their ncRNAs inside small extracellular vesicles, allowing the delivery of genetic material to neighboring or distantly located target cells. The role of these secreted RNAs in cell-to-cell communication between β-cells and other metabolic tissues as well as their potential use as diabetes biomarkers will be discussed. © 2020 American Physiological Society. Compr Physiol 10:893-932, 2020.
Collapse
Affiliation(s)
- Claudiane Guay
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Cécile Jacovetti
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Mustafa Bilal Bayazit
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Flora Brozzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Adriana Rodriguez-Trejo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Kejing Wu
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
9
|
Singh T, Suarez Castellanos I, Bhowmick DC, Cohen J, Jeremic A, Zderic V. Therapeutic Ultrasound-Induced Insulin Release in Vivo. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:639-648. [PMID: 31837888 DOI: 10.1016/j.ultrasmedbio.2019.10.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/27/2019] [Accepted: 10/27/2019] [Indexed: 06/10/2023]
Abstract
The tolerability and efficacy of low-frequency, low-intensity therapeutic ultrasound-induced insulin release was investigated in a pre-clinical in vivo murine model. The treatment groups received a single 5-min continuous sonication at 1 MHz and 1.0 W/cm2. Insulin and glucagon levels in the serum were determined using enzyme-linked immunosorbent assay. The pancreas was excised and sectioned for histologic analysis. In terminal studies, we observed a moderate (∼50 pM) but significant increase in blood insulin concentration in vivo immediately after sonication compared with a decrease of approximately 60 pM in sham animals (n < 6, p < 0.005). No difference was observed in the change in glucose or glucagon concentrations between groups. Comparisons of hematoxylin and eosin-stained terminal and survival pancreatic tissue revealed no visible differences or evidence of damage. This study is the first step in assessing the translational potential of therapeutic ultrasound as a treatment for early stages of type 2 diabetes.
Collapse
Affiliation(s)
- Tania Singh
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Ivan Suarez Castellanos
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA; INSERM, Laboratory of Therapeutic Applications of Ultrasound (LabTAU), Lyon, France
| | | | - Joshua Cohen
- Medical Faculty Associates, The George Washington University, Washington, DC, USA
| | - Aleksandar Jeremic
- Department of Biological Sciences, The George Washington University, Washington, DC, USA
| | - Vesna Zderic
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA.
| |
Collapse
|
10
|
Franzago M, Fraticelli F, Stuppia L, Vitacolonna E. Nutrigenetics, epigenetics and gestational diabetes: consequences in mother and child. Epigenetics 2019; 14:215-235. [PMID: 30865571 PMCID: PMC6557546 DOI: 10.1080/15592294.2019.1582277] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gestational Diabetes Mellitus (GDM) is the most common metabolic condition during pregnancy and may result in short- and long-term complications for both mother and offspring. The complexity of phenotypic outcomes seems influenced by genetic susceptibility, nutrient-gene interactions and lifestyle interacting with clinical factors. There is strong evidence that not only the adverse genetic background but also the epigenetic modifications in response to nutritional and environmental factors could influence the maternal hyperglycemia in pregnancy and the foetal metabolic programming. In this view, the correlation between epigenetic modifications and their transgenerational effects represents a very interesting field of study. The present review gives insight into the role of gene variants and their interactions with nutrients in GDM. In addition, we provide an overview of the epigenetic changes and their role in the maternal-foetal transmission of chronic diseases. Overall, the knowledge of epigenetic modifications induced by an adverse intrauterine and perinatal environment could shed light on the potential pathophysiological mechanisms of long-term disease development in the offspring and provide useful tools for their prevention.
Collapse
Affiliation(s)
- Marica Franzago
- a Department of Medicine and Aging, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy.,b Molecular Genetics, Unit , CeSI-Met , Chieti , Italy
| | - Federica Fraticelli
- a Department of Medicine and Aging, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy
| | - Liborio Stuppia
- b Molecular Genetics, Unit , CeSI-Met , Chieti , Italy.,c Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy
| | - Ester Vitacolonna
- a Department of Medicine and Aging, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy
| |
Collapse
|
11
|
Dusaulcy R, Handgraaf S, Visentin F, Vesin C, Philippe J, Gosmain Y. miR-132-3p is a positive regulator of alpha-cell mass and is downregulated in obese hyperglycemic mice. Mol Metab 2019; 22:84-95. [PMID: 30711402 PMCID: PMC6437597 DOI: 10.1016/j.molmet.2019.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/11/2019] [Accepted: 01/12/2019] [Indexed: 01/07/2023] Open
Abstract
Objective Diabetes is a complex disease implicating several organs and cell types. Within the islets, dysregulation occurs in both alpha- and beta-cells, leading to defects of insulin secretion and increased glucagon secretion. Dysregulation of alpha-cells is associated with transcriptome changes. We hypothesized that microRNAs (miRNAs) which are negative regulators of mRNA stability and translation could be involved in alpha-cell alterations or adaptations during type 2 diabetes. Methods miRNA microarray analyses were performed on pure alpha- and beta-cells from high-fat diet fed obese hyperglycemic mice and low-fat diet fed controls. Then, the most regulated miRNA was overexpressed or inhibited in primary culture of mouse and human alpha-cells to determine its molecular and functional impact. Results 16 miRNAs were significantly regulated in alpha-cells of obese hyperglycemic mice and 28 in beta-cells. miR-132-3p had the strongest regulation level in alpha-cells, where it was downregulated, while we observed an opposite upregulation in beta-cells. In vitro experiments showed that miR-132-3p, which is inversely regulated by somatostatin and cAMP, is a positive modulator of alpha-cell proliferation and implicated in their resistance to apoptosis. These effects are associated with the regulation of a series of genes, including proliferation and stress markers Mki67 and Bbc3 in mouse and human alpha-cells, potentially involved in miR-132-3p functions. Conclusions Downregulation of miR-132-3p in alpha-cells of obese diabetic mice may constitute a compensatory mechanism contributing to keep glucagon-producing cell number constant in diabetes. Alpha- and beta-cells present specific microRNA signatures. 16 microRNAs are significantly regulated in alpha-cells of obese hyperglycemic mice. miR-132-3p is downregulated in alpha-cells of obese hyperglycemic mice. miR-132-3p stimulates alpha-cells proliferation and resistance to apoptosis. miR-132-3p is regulated by somatostatin in alpha-cells.
Collapse
Affiliation(s)
- Rodolphe Dusaulcy
- Laboratory of Molecular Diabetes, Division of Endocrinology, Diabetes, Hypertension and Nutrition, University Hospital/Diabetes Center/University of Geneva Medical School, Geneva, Switzerland.
| | - Sandra Handgraaf
- Laboratory of Molecular Diabetes, Division of Endocrinology, Diabetes, Hypertension and Nutrition, University Hospital/Diabetes Center/University of Geneva Medical School, Geneva, Switzerland
| | - Florian Visentin
- Laboratory of Molecular Diabetes, Division of Endocrinology, Diabetes, Hypertension and Nutrition, University Hospital/Diabetes Center/University of Geneva Medical School, Geneva, Switzerland
| | - Christian Vesin
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, Geneva, Switzerland
| | - Jacques Philippe
- Laboratory of Molecular Diabetes, Division of Endocrinology, Diabetes, Hypertension and Nutrition, University Hospital/Diabetes Center/University of Geneva Medical School, Geneva, Switzerland
| | - Yvan Gosmain
- Laboratory of Molecular Diabetes, Division of Endocrinology, Diabetes, Hypertension and Nutrition, University Hospital/Diabetes Center/University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
12
|
Esguerra JLS, Nagao M, Ofori JK, Wendt A, Eliasson L. MicroRNAs in islet hormone secretion. Diabetes Obes Metab 2018; 20 Suppl 2:11-19. [PMID: 30230181 DOI: 10.1111/dom.13382] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/10/2018] [Accepted: 05/23/2018] [Indexed: 12/12/2022]
Abstract
Pancreatic islet hormone secretion is central in the maintenance of blood glucose homeostasis. During development of hyperglycaemia, the β-cell is under pressure to release more insulin to compensate for increased insulin resistance. Failure of the β-cells to secrete enough insulin results in type 2 diabetes (T2D). MicroRNAs (miRNAs) are short non-coding RNA molecules suitable for rapid regulation of the changes in target gene expression needed in β-cell adaptations. Moreover, miRNAs are involved in the maintenance of α-cell and β-cell phenotypic identities via cell-specific, or cell-enriched expression. Although many of the abundant miRNAs are highly expressed in both cell types, recent research has focused on the role of miRNAs in β-cells. It has been shown that highly abundant miRNAs, such as miR-375, are involved in several cellular functions indispensable in maintaining β-cell phenotypic identity, almost acting as "housekeeping genes" in the context of hormone secretion. Despite the abundance and importance of miR-375, it has not been shown to be differentially expressed in T2D islets. On the contrary, the less abundant miRNAs such as miR-212/miR-132, miR-335, miR-130a/b and miR-152 are deregulated in T2D islets, wherein the latter three miRNAs were shown to play key roles in regulating β-cell metabolism. In this review, we focus on β-cell function and describe miRNAs involved in insulin biosynthesis and processing, glucose uptake and metabolism, electrical activity and Ca2+ -influx and exocytosis of the insulin granules. We present current status on miRNA regulation in α-cells, and finally we discuss the involvement of miRNAs in β-cell dysfunction underlying T2D pathogenesis.
Collapse
Affiliation(s)
- Jonathan L S Esguerra
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Clinical Research Centre, SUS, Malmö, Sweden
| | - Mototsugu Nagao
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Clinical Research Centre, SUS, Malmö, Sweden
| | - Jones K Ofori
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Clinical Research Centre, SUS, Malmö, Sweden
| | - Anna Wendt
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Clinical Research Centre, SUS, Malmö, Sweden
| | - Lena Eliasson
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Clinical Research Centre, SUS, Malmö, Sweden
| |
Collapse
|
13
|
Gandasi NR, Yin P, Omar-Hmeadi M, Ottosson Laakso E, Vikman P, Barg S. Glucose-Dependent Granule Docking Limits Insulin Secretion and Is Decreased in Human Type 2 Diabetes. Cell Metab 2018; 27:470-478.e4. [PMID: 29414688 DOI: 10.1016/j.cmet.2017.12.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/26/2017] [Accepted: 12/23/2017] [Indexed: 01/19/2023]
Abstract
Glucose-stimulated insulin secretion is biphasic, with a rapid first phase and a slowly developing sustained second phase; both are disturbed in type 2 diabetes (T2D). Biphasic secretion results from vastly different release probabilities of individual insulin granules, but the morphological and molecular basis for this is unclear. Here, we show that human insulin secretion and exocytosis critically depend on the availability of membrane-docked granules and that T2D is associated with a strong reduction in granule docking. Glucose accelerated granule docking, and this effect was absent in T2D. Newly docked granules only slowly acquired release competence; this was regulated by major signaling pathways, but not glucose. Gene expression analysis indicated that key proteins involved in granule docking are downregulated in T2D, and overexpression of these proteins increased granule docking. The findings establish granule docking as an important glucose-dependent step in human insulin secretion that is dysregulated in T2D.
Collapse
Affiliation(s)
- Nikhil R Gandasi
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden
| | - Peng Yin
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden
| | - Muhmmad Omar-Hmeadi
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden
| | - Emilia Ottosson Laakso
- Diabetes and Endocrinology Unit, Department of Clinical Sciences, Lund University Diabetes Centre, 20502 Malmö, Sweden
| | - Petter Vikman
- Diabetes and Endocrinology Unit, Department of Clinical Sciences, Lund University Diabetes Centre, 20502 Malmö, Sweden
| | - Sebastian Barg
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden.
| |
Collapse
|
14
|
Rorsman P, Ashcroft FM. Pancreatic β-Cell Electrical Activity and Insulin Secretion: Of Mice and Men. Physiol Rev 2018; 98:117-214. [PMID: 29212789 PMCID: PMC5866358 DOI: 10.1152/physrev.00008.2017] [Citation(s) in RCA: 475] [Impact Index Per Article: 67.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/30/2017] [Accepted: 06/18/2017] [Indexed: 12/14/2022] Open
Abstract
The pancreatic β-cell plays a key role in glucose homeostasis by secreting insulin, the only hormone capable of lowering the blood glucose concentration. Impaired insulin secretion results in the chronic hyperglycemia that characterizes type 2 diabetes (T2DM), which currently afflicts >450 million people worldwide. The healthy β-cell acts as a glucose sensor matching its output to the circulating glucose concentration. It does so via metabolically induced changes in electrical activity, which culminate in an increase in the cytoplasmic Ca2+ concentration and initiation of Ca2+-dependent exocytosis of insulin-containing secretory granules. Here, we review recent advances in our understanding of the β-cell transcriptome, electrical activity, and insulin exocytosis. We highlight salient differences between mouse and human β-cells, provide models of how the different ion channels contribute to their electrical activity and insulin secretion, and conclude by discussing how these processes become perturbed in T2DM.
Collapse
Affiliation(s)
- Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Frances M Ashcroft
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
15
|
Zhang A, Li D, Liu Y, Li J, Zhang Y, Zhang CY. Islet β cell: An endocrine cell secreting miRNAs. Biochem Biophys Res Commun 2018; 495:1648-1654. [DOI: 10.1016/j.bbrc.2017.12.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/05/2017] [Indexed: 12/28/2022]
|
16
|
Eliasson L. The small RNA miR-375 - a pancreatic islet abundant miRNA with multiple roles in endocrine beta cell function. Mol Cell Endocrinol 2017; 456:95-101. [PMID: 28254488 DOI: 10.1016/j.mce.2017.02.043] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/20/2017] [Accepted: 02/26/2017] [Indexed: 12/21/2022]
Abstract
The pathophysiology of diabetes is complex and recent research put focus on the pancreatic islets of Langerhans and the insulin-secreting beta cells as central in the development of the disease. MicroRNAs (miRNAs), the small non-coding RNAs regulating post-transcriptional gene expression, are significant regulators of beta cell function. One of the most abundant miRNAs in the islets is miR-375. This review focus on the role of miR-375 in beta cell function, including effects in development and differentiation, proliferation and regulation of insulin secretion. It also discusses the regulation of miR-375 expression, miR-375 as a potential circulating biomarker in type 1 and type 2 diabetes, and the need for the beta cell to keep expression of miR-375 within optimal levels. The summed picture of miR-375 is a miRNA with multiple functions with importance in the formation of beta cell identity, control of beta cell mass and regulation of insulin secretion.
Collapse
Affiliation(s)
- Lena Eliasson
- Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences in Malmö, Lund University, CRC, SUS Malmö, Malmö, Sweden.
| |
Collapse
|
17
|
Hirata T, Yoshitomi T, Inoue M, Iigo Y, Matsumoto K, Kubota K, Shinagawa A. Pathological and gene expression analysis of a polygenic diabetes model, NONcNZO10/LtJ mice. Gene 2017; 629:52-58. [PMID: 28760554 DOI: 10.1016/j.gene.2017.07.075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 06/19/2017] [Accepted: 07/27/2017] [Indexed: 01/07/2023]
Abstract
The NONcNZO10/LtJ mouse is a polygenic model of type-2 diabetes (T2D) that shows moderate obesity and diabetes, and is regarded as a good model reflective of the conditions of human T2D. In this study, we analyzed pathological changes of pancreases with the progress of time by using histopathology and gene expression analysis, including microRNA. A number of gene expression changes associated with decreased insulin secretion (possibly regulated by miR-29a/b) were observed, and zinc homeostasis (Slc30a8, Mt1 and Mt2) or glucose metabolism (Slc2a2) was suggested as being the candidate mechanism of pancreas failure in NONcNZO10/LtJ mice. These results demonstrate NONcNZO10/LtJ mice have a complex pathogenic mechanism of diabetes, and moreover, this fundamental information of NONcNZO10/LtJ mice would offer the opportunity for research and development of a novel antidiabetic drug.
Collapse
Affiliation(s)
- Tsuyoshi Hirata
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13, Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Tomomi Yoshitomi
- End-Organ Disease Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Minoru Inoue
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13, Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Yutaka Iigo
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13, Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Koji Matsumoto
- End-Organ Disease Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kazuishi Kubota
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13, Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan.
| | - Akira Shinagawa
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13, Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| |
Collapse
|
18
|
Ofori JK, Malm HA, Mollet IG, Eliasson L, Esguerra JLS. Confluence does not affect the expression of miR-375 and its direct targets in rat and human insulin-secreting cell lines. PeerJ 2017; 5:e3503. [PMID: 28674658 PMCID: PMC5493031 DOI: 10.7717/peerj.3503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 06/05/2017] [Indexed: 01/01/2023] Open
Abstract
MicroRNAs are small non-coding RNAs, which negatively regulate the expression of target genes. They have emerged as important modulators in beta cell compensation upon increased metabolic demand, failure of which leads to reduced insulin secretion and type 2 diabetes. To elucidate the function of miRNAs in beta cells, insulin-secreting cell lines, such as the rat insulinoma INS-1 832/13 and the human EndoC-βH1, are widely used. Previous studies in the cancer field have suggested that miRNA expression is influenced by confluency of adherent cells. We therefore aimed to investigate whether one of the most enriched miRNAs in the pancreatic endocrine cells, miR-375, and two of its validated targets in mouse, Cav1 and Aifm1, were differentially-expressed in cell cultures with different confluences. Additionally, we measured the expression of other miRNAs, such as miR-152, miR-130a, miR-132, miR-212 and miR-200a, with known roles in beta cell function. We did not see any significant expression changes of miR-375 nor any of the two targets, in both the rat and human beta cell lines at different confluences. Interestingly, among the other miRNAs measured, the expression of miR-132 and miR-212 positively correlated with confluence, but only in the INS-1 832/13 cells. Our results show that the expression of miR-375 and other miRNAs with known roles in beta cell function is independent of, or at least minimally influenced by the density of proliferating adherent cells, especially within the confluence range optimal for functional assays to elucidate miRNA-dependent regulatory mechanisms in the beta cell.
Collapse
Affiliation(s)
- Jones K Ofori
- Department of Clinical Sciences Malmö, Lund University, SUS-Malmö, Sweden.,Lund University Diabetes Centre, Lund University, Lund and Malmö, Sweden
| | - Helena A Malm
- Department of Clinical Sciences Malmö, Lund University, SUS-Malmö, Sweden.,Lund University Diabetes Centre, Lund University, Lund and Malmö, Sweden
| | - Ines G Mollet
- Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Lena Eliasson
- Department of Clinical Sciences Malmö, Lund University, SUS-Malmö, Sweden.,Lund University Diabetes Centre, Lund University, Lund and Malmö, Sweden
| | - Jonathan Lou S Esguerra
- Department of Clinical Sciences Malmö, Lund University, SUS-Malmö, Sweden.,Lund University Diabetes Centre, Lund University, Lund and Malmö, Sweden
| |
Collapse
|
19
|
Marchetti P, Bugliani M, De Tata V, Suleiman M, Marselli L. Pancreatic Beta Cell Identity in Humans and the Role of Type 2 Diabetes. Front Cell Dev Biol 2017; 5:55. [PMID: 28589121 PMCID: PMC5440564 DOI: 10.3389/fcell.2017.00055] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/05/2017] [Indexed: 12/13/2022] Open
Abstract
Pancreatic beta cells uniquely synthetize, store, and release insulin. Specific molecular, functional as well as ultrastructural traits characterize their insulin secretion properties and survival phentoype. In this review we focus on human islet/beta cells, and describe the changes that occur in type 2 diabetes and could play roles in the disease as well as represent possible targets for therapeutical interventions. These include transcription factors, molecules involved in glucose metabolism and insulin granule handling. Quantitative and qualitative insulin release patterns and their changes in type 2 diabetes are also associated with ultrastructural features involving the insulin granules, the mitochondria, and the endoplasmic reticulum.
Collapse
Affiliation(s)
- Piero Marchetti
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Vincenzo De Tata
- Department of Translational Medicine, University of PisaPisa, Italy
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| |
Collapse
|
20
|
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: 3.8] [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
|
21
|
Sithara S, Crowley TM, Walder K, Aston-Mourney K. Gene expression signature: a powerful approach for drug discovery in diabetes. J Endocrinol 2017; 232:R131-R139. [PMID: 27927696 DOI: 10.1530/joe-16-0515] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 12/07/2016] [Indexed: 12/21/2022]
Abstract
Type 2 diabetes (T2D) is increasing in prevalence at an alarming rate around the world. Much effort has gone into the discovery and design of antidiabetic drugs; however, those already available are unable to combat the underlying causes of the disease and instead only moderate the symptoms. The reason for this is that T2D is a complex disease, and attempts to target one biological pathway are insufficient to combat the full extent of the disease. Additionally, the underlying pathophysiology of this disease is yet to be fully elucidated making it difficult to design drugs that target the mechanisms involved. Therefore, the approach of designing new drugs aimed at a specific molecular target is not optimal and a more expansive, unbiased approach is required. In this review, we will look at the current state of diabetes treatments and how these target the disease symptoms but are unable to combat the underlying causes. We will also review how the technique of gene expression signatures (GESs) has been used successfully for other complex diseases and how this may be applied as a powerful tool for the discovery of new drugs for T2D.
Collapse
Affiliation(s)
- Smithamol Sithara
- Metabolic Research UnitSchool of Medicine, Deakin University, Geelong, Australia
| | - Tamsyn M Crowley
- School of MedicineMMR, Bioinformatics Core Research Facility, Deakin University, Geelong, Australia
| | - Ken Walder
- Metabolic Research UnitSchool of Medicine, Deakin University, Geelong, Australia
| | | |
Collapse
|
22
|
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: 0.6] [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
|
23
|
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: 2.4] [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
|
24
|
Ozanne SE, Rahmoune H, Guest PC. Multiplex Biomarker Approaches in Type 2 Diabetes Mellitus Research. Methods Mol Biol 2017; 1546:37-55. [PMID: 27896756 DOI: 10.1007/978-1-4939-6730-8_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Type 2 diabetes mellitus is a multifactorial condition resulting in high fasting blood glucose levels. Although its diagnosis is straightforward, there is not one set of biomarkers or drug targets that can be used for classification or personalized treatment of individuals who suffer from this condition. Instead, the application of multiplex methods incorporating a systems biology approach is essential in order to increase our understanding of this disease. This chapter reviews the state of the art in biomarker studies of human type 2 diabetes from a proteomic and metabolomic perspective. Our main focus was on biomarkers for disease prediction as these could lead to early intervention strategies for the best possible patient outcomes.
Collapse
Affiliation(s)
- Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK. .,Department of Clinical Biochemistry, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 2QR, UK.
| | - Hassan Rahmoune
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Pembroke Street, Cambridge, UK
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
25
|
Grieco FA, Sebastiani G, Juan-Mateu J, Villate O, Marroqui L, Ladrière L, Tugay K, Regazzi R, Bugliani M, Marchetti P, Dotta F, Eizirik DL. MicroRNAs miR-23a-3p, miR-23b-3p, and miR-149-5p Regulate the Expression of Proapoptotic BH3-Only Proteins DP5 and PUMA in Human Pancreatic β-Cells. Diabetes 2017; 66:100-112. [PMID: 27737950 PMCID: PMC5204315 DOI: 10.2337/db16-0592] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 10/08/2016] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease leading to β-cell destruction. MicroRNAs (miRNAs) are small noncoding RNAs that control gene expression and organ formation. They participate in the pathogenesis of several autoimmune diseases, but the nature of miRNAs contributing to β-cell death in T1D and their target genes remain to be clarified. We performed an miRNA expression profile on human islet preparations exposed to the cytokines IL-1β plus IFN-γ. Confirmation of miRNA and target gene modification in human β-cells was performed by real-time quantitative PCR. Single-stranded miRNAs inhibitors were used to block selected endogenous miRNAs. Cell death was measured by Hoechst/propidium iodide staining and activation of caspase-3. Fifty-seven miRNAs were detected as modulated by cytokines. Three of them, namely miR-23a-3p, miR-23b-3p, and miR-149-5p, were downregulated by cytokines and selected for further studies. These miRNAs were found to regulate the expression of the proapoptotic Bcl-2 proteins DP5 and PUMA and consequent human β-cell apoptosis. These results identify a novel cross talk between a key family of miRNAs and proapoptotic Bcl-2 proteins in human pancreatic β-cells, broadening our understanding of cytokine-induced β-cell apoptosis in early T1D.
Collapse
Affiliation(s)
- Fabio Arturo Grieco
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Umberto Di Mario ONLUS Foundation-Toscana Life Sciences Foundation, Siena, Italy
| | - Jonas Juan-Mateu
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Olatz Villate
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Laura Marroqui
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Ladrière
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Ksenya Tugay
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Marco Bugliani
- Islet Cell Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Islet Cell Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Umberto Di Mario ONLUS Foundation-Toscana Life Sciences Foundation, Siena, Italy
| | - Décio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
26
|
Isik M, Blackwell TK, Berezikov E. MicroRNA mir-34 provides robustness to environmental stress response via the DAF-16 network in C. elegans. Sci Rep 2016; 6:36766. [PMID: 27905558 PMCID: PMC5131338 DOI: 10.1038/srep36766] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/20/2016] [Indexed: 12/16/2022] Open
Abstract
Diverse stresses and aging alter expression levels of microRNAs, suggesting a role for these posttranscriptional regulators of gene expression in stress modulation and longevity. Earlier studies demonstrated a central role for the miR-34 family in promoting cell cycle arrest and cell death following stress in human cells. However, the biological significance of this response was unclear. Here we show that in C. elegans mir-34 upregulation is necessary for developmental arrest, correct morphogenesis, and adaptation to a lower metabolic state to protect animals against stress-related damage. Either deletion or overexpression of mir-34 lead to an impaired stress response, which can largely be explained by perturbations in DAF-16/FOXO target gene expression. We demonstrate that mir-34 expression is regulated by the insulin signaling pathway via a negative feedback loop between miR-34 and DAF-16/FOXO. We propose that mir-34 provides robustness to stress response programs by controlling noise in the DAF-16/FOXO-regulated gene network.
Collapse
Affiliation(s)
- Meltem Isik
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands.,Joslin Diabetes Center, Harvard Stem Cell Institute and Harvard Medical School Department of Genetics, Boston, Massachusetts, United States of America
| | - T Keith Blackwell
- Joslin Diabetes Center, Harvard Stem Cell Institute and Harvard Medical School Department of Genetics, Boston, Massachusetts, United States of America
| | - Eugene Berezikov
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands.,European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
27
|
Mollet IG, Malm HA, Wendt A, Orho-Melander M, Eliasson L. Integrator of Stress Responses Calmodulin Binding Transcription Activator 1 (Camta1) Regulates miR-212/miR-132 Expression and Insulin Secretion. J Biol Chem 2016; 291:18440-52. [PMID: 27402838 DOI: 10.1074/jbc.m116.716860] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Indexed: 01/04/2023] Open
Abstract
Altered microRNA profiles have been demonstrated in experimental models of type 2 diabetes, including in islets of the diabetic Goto-Kakizaki (GK) rat. Our bioinformatic analysis of conserved sequences in promoters of microRNAs, previously observed to be up-regulated in GK rat islets, revealed putative CGCG-core motifs on the promoter of the miR-212/miR-132 cluster, overexpression of which has been shown to increase insulin secretion. These motifs are possible targets of calmodulin binding transcription activators Camta1 and Camta2 that have been recognized as integrators of stress responses. We also identified putative NKE elements, possible targets of NK2 homeobox proteins like the essential islet transcription factor Nkx2-2. As Camtas can function as co-activators with NK2 proteins in other tissues, we explored the role of Camta1, Camta2, and Nkx2-2 in the regulation of the miR-212/miR-132 cluster and insulin secretion. We demonstrate that exposure of control Wistar or GK rat islets to 16.7 mm glucose increases miR-212/miR-132 expression but significantly less so in the GK rat. In addition, Camta1, Camta2, and Nkx2-2 were down-regulated in GK rat islets, and knockdown of Camta1 reduced miR-212/miR-132 promoter activity and miR-212/miR-132 expression, even under cAMP elevation. Knockdown of Camta1 decreased insulin secretion in INS-1 832/13 cells and Wistar rat islets but increased insulin content. Furthermore, knockdown of Camta1 reduced K(+)-induced insulin secretion and voltage-dependent Ca(2+) currents. We also demonstrate Camta1 and Nkx2-2 protein interaction. These results indicate that Camta1 is required not only for expression of the miR-212/miR-132 cluster but at multiple levels for regulating beta cell insulin content and secretion.
Collapse
Affiliation(s)
- Inês Guerra Mollet
- From the Department of Clinical Sciences, Clinical Research Centre, Lund University, Jan Waldenströms Gata 35, SE-20502 Malmö, Sweden
| | - Helena Anna Malm
- From the Department of Clinical Sciences, Clinical Research Centre, Lund University, Jan Waldenströms Gata 35, SE-20502 Malmö, Sweden
| | - Anna Wendt
- From the Department of Clinical Sciences, Clinical Research Centre, Lund University, Jan Waldenströms Gata 35, SE-20502 Malmö, Sweden
| | - Marju Orho-Melander
- From the Department of Clinical Sciences, Clinical Research Centre, Lund University, Jan Waldenströms Gata 35, SE-20502 Malmö, Sweden
| | - Lena Eliasson
- From the Department of Clinical Sciences, Clinical Research Centre, Lund University, Jan Waldenströms Gata 35, SE-20502 Malmö, Sweden
| |
Collapse
|
28
|
Miller DB, Snow SJ, Henriquez A, Schladweiler MC, Ledbetter AD, Richards JE, Andrews DL, Kodavanti UP. Systemic metabolic derangement, pulmonary effects, and insulin insufficiency following subchronic ozone exposure in rats. Toxicol Appl Pharmacol 2016; 306:47-57. [PMID: 27368153 DOI: 10.1016/j.taap.2016.06.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/23/2016] [Accepted: 06/26/2016] [Indexed: 12/17/2022]
Abstract
Acute ozone exposure induces a classical stress response with elevated circulating stress hormones along with changes in glucose, protein and lipid metabolism in rats, with similar alterations in ozone-exposed humans. These stress-mediated changes over time have been linked to insulin resistance. We hypothesized that acute ozone-induced stress response and metabolic impairment would persist during subchronic episodic exposure and induce peripheral insulin resistance. Male Wistar Kyoto rats were exposed to air or 0.25ppm or 1.00ppm ozone, 5h/day, 3 consecutive days/week (wk) for 13wks. Pulmonary, metabolic, insulin signaling and stress endpoints were determined immediately after 13wk or following a 1wk recovery period (13wk+1wk recovery). We show that episodic ozone exposure is associated with persistent pulmonary injury and inflammation, fasting hyperglycemia, glucose intolerance, as well as, elevated circulating adrenaline and cholesterol when measured at 13wk, however, these responses were largely reversible following a 1wk recovery. Moreover, the increases noted acutely after ozone exposure in non-esterified fatty acids and branched chain amino acid levels were not apparent following a subchronic exposure. Neither peripheral or tissue specific insulin resistance nor increased hepatic gluconeogenesis were present after subchronic ozone exposure. Instead, long-term ozone exposure lowered circulating insulin and severely impaired glucose-stimulated beta-cell insulin secretion. Thus, our findings in young-adult rats provide potential insights into epidemiological studies that show a positive association between ozone exposures and type 1 diabetes. Ozone-induced beta-cell dysfunction may secondarily contribute to other tissue-specific metabolic alterations following chronic exposure due to impaired regulation of glucose, lipid, and protein metabolism.
Collapse
Affiliation(s)
- Desinia B Miller
- Curriculum in Toxicology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, United States
| | - Samantha J Snow
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - Andres Henriquez
- Curriculum in Toxicology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, United States
| | - Mette C Schladweiler
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - Allen D Ledbetter
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - Judy E Richards
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - Debora L Andrews
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - Urmila P Kodavanti
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States.
| |
Collapse
|
29
|
Bavia L, Mosimann ALP, Aoki MN, Duarte Dos Santos CN. A glance at subgenomic flavivirus RNAs and microRNAs in flavivirus infections. Virol J 2016; 13:84. [PMID: 27233361 PMCID: PMC4884392 DOI: 10.1186/s12985-016-0541-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/17/2016] [Indexed: 11/10/2022] Open
Abstract
The family Flaviviridae comprises a wide variety of viruses that are distributed worldwide, some of which are associated with high rates of morbidity and mortality. There are neither vaccines nor antivirals for most flavivirus infections, reinforcing the importance of research on different aspects of the viral life cycle. During infection, cytoplasmic accumulation of RNA fragments mainly originating from the 3' UTRs, which have been designated subgenomic flavivirus RNAs (sfRNAs), has been detected. It has been shown that eukaryotic exoribonucleases are involved in viral sfRNA production. Additionally, viral and human small RNAs (sRNAs) have also been found in flavivirus-infected cells, especially microRNAs (miRNAs). miRNAs were first described in eukaryotic cells and in a mature and functional state present as single-stranded 18-24 nt RNA fragments. Their main function is the repression of translation through base pairing with cellular mRNAs, besides other functions, such as mRNA degradation. Canonical miRNA biogenesis involves Drosha and Dicer, however miRNA can also be generated by alternative pathways. In the case of flaviviruses, alternative pathways have been suggested. Both sfRNAs and miRNAs are involved in viral infection and host cell response modulation, representing interesting targets of antiviral strategies. In this review, we focus on the generation and function of viral sfRNAs, sRNAs and miRNAs in West Nile, dengue, Japanese encephalitis, Murray Valley encephalitis and yellow fever infections, as well as their roles in viral replication, translation and cell immune response evasion. We also give an overview regarding other flaviviruses and the generation of cellular miRNAs during infection.
Collapse
Affiliation(s)
- Lorena Bavia
- Laboratório de Virologia Molecular, Instituto Carlos Chagas (ICC/FIOCRUZ-PR), Rua Prof. Algacyr Munhoz Mader 3775, CIC, CEP: 81350-010, Curitiba, Paraná, Brazil
| | - Ana Luiza Pamplona Mosimann
- Laboratório de Virologia Molecular, Instituto Carlos Chagas (ICC/FIOCRUZ-PR), Rua Prof. Algacyr Munhoz Mader 3775, CIC, CEP: 81350-010, Curitiba, Paraná, Brazil
| | - Mateus Nóbrega Aoki
- Laboratório de Virologia Molecular, Instituto Carlos Chagas (ICC/FIOCRUZ-PR), Rua Prof. Algacyr Munhoz Mader 3775, CIC, CEP: 81350-010, Curitiba, Paraná, Brazil
| | - Claudia Nunes Duarte Dos Santos
- Laboratório de Virologia Molecular, Instituto Carlos Chagas (ICC/FIOCRUZ-PR), Rua Prof. Algacyr Munhoz Mader 3775, CIC, CEP: 81350-010, Curitiba, Paraná, Brazil.
| |
Collapse
|
30
|
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: 55] [Impact Index Per Article: 6.1] [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
|
31
|
Salazar J, Luzardo E, Mejías JC, Rojas J, Ferreira A, Rivas-Ríos JR, Bermúdez V. Epicardial Fat: Physiological, Pathological, and Therapeutic Implications. Cardiol Res Pract 2016; 2016:1291537. [PMID: 27213076 PMCID: PMC4861775 DOI: 10.1155/2016/1291537] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 03/02/2016] [Accepted: 04/03/2016] [Indexed: 12/20/2022] Open
Abstract
Epicardial fat is closely related to blood supply vessels, both anatomically and functionally, which is why any change in this adipose tissue's behavior is considered a potential risk factor for cardiovascular disease development. When proinflammatory adipokines are released from the epicardial fat, this can lead to a decrease in insulin sensitivity, low adiponectin production, and an increased proliferation of vascular smooth muscle cells. These adipokines move from one compartment to another by either transcellular passing or diffusion, thus having the ability to regulate cardiac muscle activity, a phenomenon called vasocrine regulation. The participation of these adipokines generates a state of persistent vasoconstriction, increased stiffness, and weakening of the coronary wall, consequently contributing to the formation of atherosclerotic plaques. Therefore, epicardial adipose tissue thickening should be considered a risk factor in the development of cardiovascular disease, a potential therapeutic target for cardiovascular pathology and a molecular point of contact for "endocrine-cardiology."
Collapse
Affiliation(s)
- Juan Salazar
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
| | - Eliana Luzardo
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
| | - José Carlos Mejías
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
| | - Joselyn Rojas
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Antonio Ferreira
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
- Internal Medicine Service, “Dr. Manuel Noriega Trigo” Hospital, San Francisco 4004, Venezuela
| | - José Ramón Rivas-Ríos
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
| | - Valmore Bermúdez
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
| |
Collapse
|
32
|
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: 4.7] [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
|
33
|
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: 2.2] [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]
|
34
|
Abstract
Insulin is a key hormone controlling metabolic homeostasis. Loss or dysfunction of pancreatic β-cells lead to the release of insufficient insulin to cover the organism needs, promoting diabetes development. Since dietary nutrients influence the activity of β-cells, their inadequate intake, absorption and/or utilisation can be detrimental. This review will highlight the physiological and pathological effects of nutrients on insulin secretion and discuss the underlying mechanisms. Glucose uptake and metabolism in β-cells trigger insulin secretion. This effect of glucose is potentiated by amino acids and fatty acids, as well as by entero-endocrine hormones and neuropeptides released by the digestive tract in response to nutrients. Glucose controls also basal and compensatory β-cell proliferation and, along with fatty acids, regulates insulin biosynthesis. If in the short-term nutrients promote β-cell activities, chronic exposure to nutrients can be detrimental to β-cells and causes reduced insulin transcription, increased basal secretion and impaired insulin release in response to stimulatory glucose concentrations, with a consequent increase in diabetes risk. Likewise, suboptimal early-life nutrition (e.g. parental high-fat or low-protein diet) causes altered β-cell mass and function in adulthood. The mechanisms mediating nutrient-induced β-cell dysfunction include transcriptional, post-transcriptional and translational modifications of genes involved in insulin biosynthesis and secretion, carbohydrate and lipid metabolism, cell differentiation, proliferation and survival. Altered expression of these genes is partly caused by changes in non-coding RNA transcripts induced by unbalanced nutrient uptake. A better understanding of the mechanisms leading to β-cell dysfunction will be critical to improve treatment and find a cure for diabetes.
Collapse
|
35
|
Bhatia P, Raina S, Chugh J, Sharma S. miRNAs: early prognostic biomarkers for Type 2 diabetes mellitus? Biomark Med 2015; 9:1025-40. [DOI: 10.2217/bmm.15.69] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) has reached epidemic proportions and is associated with peripheral insulin resistance. The currently used therapies aim to delay progression of T2DM. Their efficacy could drastically be improved if implemented at earlier stages. Classical diagnostic markers (blood glucose and HbA1C) are generally detected once metabolic imbalance has already set in. Therefore, development of biomarkers for early diagnosis would help identify individuals at risk for developing T2DM. Along with genetic predisposition, epigenetics also plays a major role in T2DM development. In this review, we discuss the potential role of early diagnostic markers such as circulating miRNAs, studies done so far and challenges to be considered while taking into account the novel role of miRNAs as prognostic biomarkers.
Collapse
Affiliation(s)
- Parnika Bhatia
- Department of Biotechnology, Savitribai Phule Pune University (Formerly University of Pune), Ganeshkhind, Pune 411007, India
| | - Shikha Raina
- Department of Biotechnology, Savitribai Phule Pune University (Formerly University of Pune), Ganeshkhind, Pune 411007, India
| | - Jeetender Chugh
- Department of Chemistry, Indian Institute of Science Education & Research, Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Shilpy Sharma
- Department of Biotechnology, Savitribai Phule Pune University (Formerly University of Pune), Ganeshkhind, Pune 411007, India
| |
Collapse
|
36
|
Gillberg L, Ling C. The potential use of DNA methylation biomarkers to identify risk and progression of type 2 diabetes. Front Endocrinol (Lausanne) 2015; 6:43. [PMID: 25870586 PMCID: PMC4378313 DOI: 10.3389/fendo.2015.00043] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 03/11/2015] [Indexed: 12/29/2022] Open
Abstract
Type 2 diabetes mellitus (T2D) is a slowly progressive disease that can be postponed or even avoided through lifestyle changes. Recent data demonstrate highly significant correlations between DNA methylation and the most important risk factors of T2D, including age and body mass index, in blood and human tissues relevant to insulin resistance and T2D. Also, T2D patients and individuals with increased risk of the disease display differential DNA methylation profiles and plasticity compared to controls. Accordingly, the novel clues to DNA methylation fingerprints in blood and tissues with deteriorated metabolic capacity indicate that blood-borne epigenetic biomarkers of T2D progression might become a reality. This Review will address the most recent associations between DNA methylation and diabetes-related traits in human tissues and blood. The overall focus is on the potential of future epigenome-wide studies, carried out across tissues and populations with correlations to pre-diabetes and T2D risk factors, to build up a library of epigenetic markers of risk and early progression of T2D. These markers may, tentatively in combination with other predictors of T2D development, increase the possibility of individual-based lifestyle prevention of T2D and associated metabolic diseases.
Collapse
Affiliation(s)
- Linn Gillberg
- Diabetes and Metabolism, Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Linn Gillberg, Diabetes and Metabolism, Department of Endocrinology, Rigshospitalet, Tagensvej 20, Section 7652, Copenhagen, DK-2200, Denmark e-mail:
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Malmö, Sweden
| |
Collapse
|