1
|
Zhang Y, Qian B, Yang Y, Niu F, Lin C, Yuan H, Wang J, Wu T, Shao Y, Shao S, Liu A, Wu J, Sun P, Chang X, Bi Y, Tang W, Zhu Y, Chen F, Su D, Han X. Visceral Adipocyte-Derived Extracellular Vesicle miR-27a-5p Elicits Glucose Intolerance by Inhibiting Pancreatic β-Cell Insulin Secretion. Diabetes 2024; 73:1832-1847. [PMID: 39186314 PMCID: PMC11493764 DOI: 10.2337/db24-0177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 08/09/2024] [Indexed: 08/27/2024]
Abstract
Pancreatic β-cell dysfunction caused by obesity can be associated with alterations in the levels of miRNAs. However, the role of miRNAs in such processes remains elusive. Here, we show that pancreatic islet miR-27a-5p, which is markedly increased in obese mice and impairs insulin secretion, is mainly delivered by visceral adipocyte-derived extracellular vesicles (EVs). Depleting miR-27a-5p significantly improved insulin secretion and glucose intolerance in db/db mice. Supporting the function of EV miR-27a-5p as a key pathogenic factor, intravenous injection of miR-27a-5p-containing EVs showed their distribution in mouse pancreatic islets. Tracing the injected adeno-associated virus (AAV)-miR-27a-5p (AAV-miR-27a) or AAV-FABP4-miR-27a-5p (AAV-FABP4-miR-27a) in visceral fat resulted in upregulating miR-27a-5p in EVs and serum and elicited mouse pancreatic β-cell dysfunction. Mechanistically, miR-27a-5p directly targeted L-type Ca2+ channel subtype CaV1.2 (Cacna1c) and reduced insulin secretion in β-cells. Overexpressing mouse CaV1.2 largely abolished the insulin secretion injury induced by miR-27a-5p. These findings reveal a causative role of EV miR-27a-5p in visceral adipocyte-mediated pancreatic β-cell dysfunction in obesity-associated type 2 diabetes mellitus. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yaqin Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bin Qian
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yang Yang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Fandi Niu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, Xi’an, Shanxi, China
| | - Changsong Lin
- Department of Bioinformatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Honglei Yuan
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianan Wang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tijun Wu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yixue Shao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shulin Shao
- Department of Laboratory, Nanjing Pukou Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Aiming Liu
- The First Clinical School of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingwen Wu
- The First Clinical School of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peng Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital affiliated to Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, Jiangsu, China
| | - Wei Tang
- Department of Endocrinology, Islet Cell Senescence and Function Research Laboratory, Jiangsu Province Geriatric Institute, Nanjing, Jiangsu, China
| | - Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fang Chen
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongming Su
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Yuan H, He M, Yang Q, Niu F, Zou Y, Liu C, Yang Yang, Liu A, Chang X, Chen F, Wu T, Han X, Zhang Y. Obesity-induced upregulation of miR-483-5p impairs the function and identity of pancreatic β-cells. Diabetes Obes Metab 2024; 26:4510-4521. [PMID: 39072950 DOI: 10.1111/dom.15805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/15/2024] [Accepted: 06/28/2024] [Indexed: 07/30/2024]
Abstract
AIM To assess the expression and function of miR-483-5p in diabetic β cells. METHODS The expression of miR-483-5p was evaluated in the pancreatic islets of obesity mouse models by quantitative reverse transcription polymerase chain reaction. Dual-luciferase activity, and western blotting assays, were utilized for miR-483-5p target gene verification. Mice with β cell-specific miR-483-5p downregulation were studied under metabolic stress (i.e. a high-fat diet) condition. Lineage tracing was used to determine β-cell fate. RESULTS miR-483-5p increased in the islets of obese mouse models. Expression levels of miR-483-5p were significantly upregulated with the treatment of high glucose and palmitate, in both MIN6 cells and mouse islets. Overexpression of miR-483-5p in β cells results in impaired insulin secretion and β-cell identity. Cell lineage-specific analyses revealed that miR-483-5p overexpression deactivated β-cell identity genes (insulin, Pdx1 and MafA) and derepressed β-cell dedifferentiation (Ngn3) genes. miR-483-5p downregulation in β cells of high-fat diet-fed mice alleviated diabetes and improved glucose intolerance by enhancing insulin secretory capacity. These detrimental effects of miR-483-5p relied on its seed sequence recognition and repressed expression of its target genes Pdx1 and MafA, two crucial markers of β-cell maturation. CONCLUSIONS These findings indicate that the miR-483-5p-mediated reduction of mRNAs specifies β-cell identity as a contributor to β-cell dysfunction via the loss of cellular differentiation.
Collapse
Affiliation(s)
- Honglei Yuan
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Mei He
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Qinnan Yang
- The First Clinical School of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Fandi Niu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Yuchen Zou
- The First Clinical School of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Chen Liu
- The First Clinical School of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yang Yang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Aiming Liu
- The First Clinical School of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Fang Chen
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Tijun Wu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Yaqin Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Sandovici I, Fernandez-Twinn DS, Campbell N, Cooper WN, Sekita Y, Zvetkova I, Ferland-McCollough D, Prosser HM, Oyama LM, Pantaleão LC, Cimadomo D, Barbosa de Queiroz K, Cheuk CSK, Smith NM, Kay RG, Antrobus R, Hoelle K, Ma MKL, Smith NH, Geyer SH, Reissig LF, Weninger WJ, Siddle K, Willis AE, Lam BYH, Bushell M, Ozanne SE, Constância M. Overexpression of Igf2-derived Mir483 inhibits Igf1 expression and leads to developmental growth restriction and metabolic dysfunction in mice. Cell Rep 2024; 43:114750. [PMID: 39283743 DOI: 10.1016/j.celrep.2024.114750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/04/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024] Open
Abstract
Mir483 is a conserved and highly expressed microRNA in placental mammals, embedded within the Igf2 gene. Its expression is dysregulated in a number of human diseases, including metabolic disorders and certain cancers. Here, we investigate the developmental regulation and function of Mir483 in vivo. We find that Mir483 expression is dependent on Igf2 transcription and the regulation of the Igf2/H19 imprinting control region. Transgenic Mir483 overexpression in utero causes fetal, but not placental, growth restriction through insulin-like growth factor 1 (IGF1) and IGF2 and also causes cardiovascular defects leading to fetal death. Overexpression of Mir483 post-natally results in growth stunting through IGF1 repression, increased hepatic lipid production, and excessive adiposity. IGF1 infusion rescues the post-natal growth restriction. Our findings provide insights into the function of Mir483 as a growth suppressor and metabolic regulator and suggest that it evolved within the INS-IGF2-H19 transcriptional region to limit excessive tissue growth through repression of IGF signaling.
Collapse
Affiliation(s)
- Ionel Sandovici
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Denise S Fernandez-Twinn
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Niamh Campbell
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Wendy N Cooper
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Yoichi Sekita
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Ilona Zvetkova
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | | | - Haydn M Prosser
- The Wellcome Trust Sanger Institute, Genome Campus, Hinxton, UK
| | - Lila M Oyama
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Departmento de Fisiologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil
| | - Lucas C Pantaleão
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Danilo Cimadomo
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Laboratory of Developmental Biology, Department of Biology and Biotechnology "Lazzaro Spallanzani," University of Pavia, Pavia, Italy
| | - Karina Barbosa de Queiroz
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Cecilia S K Cheuk
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Nicola M Smith
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Richard G Kay
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Katharina Hoelle
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Marcella K L Ma
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Noel H Smith
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Stefan H Geyer
- Center for Anatomy and Cell Biology, Division of Anatomy, Medical University of Vienna, Vienna, Austria
| | - Lukas F Reissig
- Center for Anatomy and Cell Biology, Division of Anatomy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang J Weninger
- Center for Anatomy and Cell Biology, Division of Anatomy, Medical University of Vienna, Vienna, Austria
| | - Kenneth Siddle
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Anne E Willis
- Medical Research Council Toxicology Unit, University of Leicester, Leicester, UK
| | - Brian Y H Lam
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Martin Bushell
- Medical Research Council Toxicology Unit, University of Leicester, Leicester, UK
| | - Susan E Ozanne
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Miguel Constância
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
4
|
Ghasemi Gojani E, Rai S, Norouzkhani F, Shujat S, Wang B, Li D, Kovalchuk O, Kovalchuk I. Targeting β-Cell Plasticity: A Promising Approach for Diabetes Treatment. Curr Issues Mol Biol 2024; 46:7621-7667. [PMID: 39057094 PMCID: PMC11275945 DOI: 10.3390/cimb46070453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The β-cells within the pancreas play a pivotal role in insulin production and secretion, responding to fluctuations in blood glucose levels. However, factors like obesity, dietary habits, and prolonged insulin resistance can compromise β-cell function, contributing to the development of Type 2 Diabetes (T2D). A critical aspect of this dysfunction involves β-cell dedifferentiation and transdifferentiation, wherein these cells lose their specialized characteristics and adopt different identities, notably transitioning towards progenitor or other pancreatic cell types like α-cells. This process significantly contributes to β-cell malfunction and the progression of T2D, often surpassing the impact of outright β-cell loss. Alterations in the expressions of specific genes and transcription factors unique to β-cells, along with epigenetic modifications and environmental factors such as inflammation, oxidative stress, and mitochondrial dysfunction, underpin the occurrence of β-cell dedifferentiation and the onset of T2D. Recent research underscores the potential therapeutic value for targeting β-cell dedifferentiation to manage T2D effectively. In this review, we aim to dissect the intricate mechanisms governing β-cell dedifferentiation and explore the therapeutic avenues stemming from these insights.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.)
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.)
| |
Collapse
|
5
|
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
|
6
|
Jiao P, Lu H, Hao L, Degen AA, Cheng J, Yin Z, Mao S, Xue Y. Nutrigenetic and Epigenetic Mechanisms of Maternal Nutrition-Induced Glucolipid Metabolism Changes in the Offspring. Nutr Rev 2024:nuae048. [PMID: 38781288 DOI: 10.1093/nutrit/nuae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Maternal nutrition during pregnancy regulates the offspring's metabolic homeostasis, including insulin sensitivity and the metabolism of glucose and lipids. The fetus undergoes a crucial period of plasticity in the uterus; metabolic changes in the fetus during pregnancy caused by maternal nutrition not only influence fetal growth and development but also have a long-term or even life-long impact for the offspring. Epigenetic modifications, such as DNA methylation, histone modification, and non-coding RNAs, play important roles in intergenerational and transgenerational effects. In this context, this narrative review comprehensively summarizes and analyzes the molecular mechanisms underlying how maternal nutrition, including a high-fat diet, polyunsaturated fatty acid diet, methyl donor nutrient supplementation, feed restriction, and protein restriction during pregnancy, impacts the genes involved in glucolipid metabolism in the liver, adipose tissue, hypothalamus, muscle, and oocytes of the offspring in terms of the epigenetic modifications. This will provide a foundation for the further exploration of nutrigenetic and epigenetic mechanisms for integrative mother-child nutrition and promotion of the offspring's health through the regulation of maternal nutrition during pregnancy. Note: This paper is part of the Nutrition Reviews Special Collection on Precision Nutrition.
Collapse
Affiliation(s)
- Peng Jiao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Huizhen Lu
- Biotechnology Center, Anhui Agricultural University, Hefei, China
| | - Lizhuang Hao
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Plateau Yak Research Center, Qinghai Academy of Science and Veterinary Medicine of Qinghai University, Xining, China
| | - A Allan Degen
- Desert Animal Adaptations and Husbandry, Wyler Department of Dryland Agriculture, Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jianbo Cheng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Shengyong Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yanfeng Xue
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
7
|
Wander PL, Bammler TK, MacDonald JW, Srinouanprachanh S, Boyko EJ, Enquobahrie DA. Plasma miRNAs and Treatment Failure in Participants in the Treatment Options for Type 2 Diabetes in Adolescents and Youth (TODAY) Study. Diabetes Care 2024; 47:471-475. [PMID: 38153877 PMCID: PMC10909680 DOI: 10.2337/dc23-1540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/15/2023] [Indexed: 12/30/2023]
Abstract
OBJECTIVE To identify plasma miRNAs related to treatment failure in youth with type 2 diabetes (T2D). RESEARCH DESIGN AND METHODS We examined whether a panel of miRNAs could predict treatment failure in training/test data sets among participants in the Treatment Options for Type 2 Diabetes in Adolescents and Youth (TODAY) study (N = 209). We also examined whether individual miRNAs were associated with treatment failure. RESULTS Participants were age 14.5 years, and 62% were female. A panel of miRNAs did not predict treatment failure. However, for each doubling, miR-4306 was associated with a 12% decrease (P = 0.040) and miR-483-3p was marginally associated with a 12% increase (P = 0.080) in failure independently of sex, race/ethnicity, BMI, Tanner stage, HbA1c, maternal diabetes, oral disposition index, and treatment arm. The addition of both miRNAs improved model fit (log likelihood without vs. with miRNAs -360.3 vs. -363.5; P = 0.040). CONCLUSIONS miR-483-3p and miR-4306 may be associated with treatment failure in youth with T2D.
Collapse
Affiliation(s)
- Pandora L. Wander
- Veterans Affairs Puget Sound Health Care System, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Theo K. Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - James W. MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Sengeo Srinouanprachanh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Edward J. Boyko
- Veterans Affairs Puget Sound Health Care System, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Daniel A. Enquobahrie
- Department of Epidemiology, University of Washington, Seattle, WA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA
| |
Collapse
|
8
|
Abulsoud AI, Elshaer SS, Abdelmaksoud NM, Zaki MB, El-Mahdy HA, Ismail A, Al-Noshokaty TM, Fathi D, Abdel-Reheim MA, Mohammed OA, Doghish AS. Investigating the regulatory role of miRNAs as silent conductors in the management of pathogenesis and therapeutic resistance of pancreatic cancer. Pathol Res Pract 2023; 251:154855. [PMID: 37806169 DOI: 10.1016/j.prp.2023.154855] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 09/16/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
Pancreatic cancer (PC) has the greatest mortality rate of all the main malignancies. Its advanced stage and poor prognosis place it at the bottom of all cancer sites. Hence, emerging biomarkers can enable precision medicine where PC therapy is tailored to each patient. This highlights the need for new, highly sensitive and specific biomarkers for early PC diagnosis. Prognostic indicators are also required to stratify PC patients. To avoid ineffective treatment, adverse events, and expenses, biomarkers are also required for patient monitoring and identifying responders to treatment. There is substantial evidence that microRNAs (miRs, miRNAs) play a critical role in regulating mRNA and, as a consequence, protein expression in normal and malignant tissues. Deregulated miRNA profiling in PC can help with diagnosis, treatment planning, and prognosis. Furthermore, knowledge of the primary effector genes and downstream pathways in PC can help pinpoint potential miRNAs for use in treatment. Different miRNA expression profiles may serve as diagnostic, prognostic markers, and therapeutic targets across the spectrum of malignant pancreatic illness. Dysregulation of miRNAs has been linked to the malignant pathophysiology of PC through affecting many cellular functions such as increasing invasive and proliferative prospect, supporting angiogenesis, cell cycle aberrance, apoptosis elusion, metastasis promotion, and low sensitivity to particular treatments. Accordingly, in the current review, we summarize the recent advances in the roles of oncogenic and tumor suppressor (TS) miRNAs in PC and discuss their potential as worthy diagnostic and prognostic biomarkers for PC, as well as their significance in PC pathogenesis and anticancer drug resistance.
Collapse
Affiliation(s)
- Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt; Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Shereen Saeid Elshaer
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt; Department of Biochemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr city, Cairo 11823, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt
| | - Tohada M Al-Noshokaty
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Doaa Fathi
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Osama A Mohammed
- Department of Clinical Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| |
Collapse
|
9
|
Afsharmanesh MR, Mohammadi Z, Mansourian AR, Jafari SM. A Review of micro RNAs changes in T2DM in animals and humans. J Diabetes 2023; 15:649-664. [PMID: 37329278 PMCID: PMC10415875 DOI: 10.1111/1753-0407.13431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 04/22/2023] [Accepted: 05/24/2023] [Indexed: 06/19/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) and its associated complications have become a crucial public health concern in the world. According to the literature, chronic inflammation and the progression of T2DM have a close relationship. Accumulated evidence suggests that inflammation enhances the insulin secretion lost by islets of Langerhans and the resistance of target tissues to insulin action, which are two critical features in T2DM development. Based on recently highlighted research that plasma concentration of inflammatory mediators such as tumor necrosis factor α and interleukin-6 are elevated in insulin-resistant and T2DM, and it raises novel question marks about the processes causing inflammation in both situations. Over the past few decades, microRNAs (miRNAs), a class of short, noncoding RNA molecules, have been discovered to be involved in the regulation of inflammation, insulin resistance, and T2DM pathology. These noncoding RNAs are specifically comprised of RNA-induced silencing complexes and regulate the expression of specific protein-coding genes through various mechanisms. There is extending evidence that describes the expression profile of a special class of miRNA molecules altered during T2DM development. These modifications can be observed as potential biomarkers for the diagnosis of T2DM and related diseases. In this review study, after reviewing the possible mechanisms involved in T2DM pathophysiology, we update recent information on the miRNA roles in T2DM, inflammation, and insulin resistance.
Collapse
Affiliation(s)
- Mohammad Reza Afsharmanesh
- Metabolic Disorders Research CenterGolestan University of Medical SciencesGorganIran
- Department of Biochemistry and Biophysics, School of MedicineGolestan University of Medical SciencesGorganIran
| | - Zeinab Mohammadi
- Metabolic Disorders Research CenterGolestan University of Medical SciencesGorganIran
- Department of Biochemistry and Biophysics, School of MedicineGolestan University of Medical SciencesGorganIran
| | - Azad Reza Mansourian
- Metabolic Disorders Research CenterGolestan University of Medical SciencesGorganIran
- Department of Biochemistry and Biophysics, School of MedicineGolestan University of Medical SciencesGorganIran
| | - Seyyed Mehdi Jafari
- Metabolic Disorders Research CenterGolestan University of Medical SciencesGorganIran
- Department of Biochemistry and Biophysics, School of MedicineGolestan University of Medical SciencesGorganIran
| |
Collapse
|
10
|
Matson K, Macleod A, Mehta N, Sempek E, Tang X. Impacts of MicroRNA-483 on Human Diseases. Noncoding RNA 2023; 9:37. [PMID: 37489457 PMCID: PMC10366739 DOI: 10.3390/ncrna9040037] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/13/2023] [Accepted: 06/21/2023] [Indexed: 07/26/2023] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNA molecules that regulate gene expression by targeting specific messenger RNAs (mRNAs) in distinct cell types. This review provides a com-prehensive overview of the current understanding regarding the involvement of miR-483-5p and miR-483-3p in various physiological and pathological processes. Downregulation of miR-483-5p has been linked to numerous diseases, including type 2 diabetes, fatty liver disease, diabetic nephropathy, and neurological injury. Accumulating evidence indicates that miR-483-5p plays a crucial protective role in preserving cell function and viability by targeting specific transcripts. Notably, elevated levels of miR-483-5p in the bloodstream strongly correlate with metabolic risk factors and serve as promising diagnostic markers. Consequently, miR-483-5p represents an appealing biomarker for predicting the risk of developing diabetes and cardiovascular diseases and holds potential as a therapeutic target for intervention strategies. Conversely, miR-483-3p exhibits significant upregulation in diabetes and cardiovascular diseases and has been shown to induce cellular apoptosis and lipotoxicity across various cell types. However, some discrepancies regarding its precise function have been reported, underscoring the need for further investigation in this area.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoqing Tang
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931, USA; (K.M.); (A.M.); (N.M.); (E.S.)
| |
Collapse
|
11
|
Volodko O, Volinsky N, Yarkoni M, Margalit N, Kusniec F, Sudarsky D, Elbaz-Greener G, Carasso S, Amir O. Characterization of Systemic and Culprit-Coronary Artery miR-483-5p Expression in Chronic CAD and Acute Myocardial Infarction Male Patients. Int J Mol Sci 2023; 24:ijms24108551. [PMID: 37239897 DOI: 10.3390/ijms24108551] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Coronary artery disease (CAD) is the leading cause of mortality worldwide. In chronic and myocardial infarction (MI) states, aberrant levels of circulating microRNAs compromise gene expression and pathophysiology. We aimed to compare microRNA expression in chronic-CAD and acute-MI male patients in peripheral blood vasculature versus coronary arteries proximal to a culprit area. Blood from chronic-CAD, acute-MI with/out ST segment elevation (STEMI/NSTEMI, respectively), and control patients lacking previous CAD or having patent coronary arteries was collected during coronary catheterization from peripheral arteries and from proximal culprit coronary arteries aimed for the interventions. Random coronary arterial blood was collected from controls; RNA extraction, miRNA library preparation and Next Generation Sequencing followed. High concentrations of microRNA-483-5p (miR-483-5p) were noted as 'coronary arterial gradient' in culprit acute-MI versus chronic-CAD (p = 0.035) which were similar to controls versus chronic-CAD (p < 0.001). Meanwhile, peripheral miR-483-5p was downregulated in acute-MI and chronic-CAD, compared with controls (1.1 ± 2.2 vs. 2.6 ± 3.3, respectively, p < 0.005). A receiver operating characteristic curve analysis for miR483-5p association with chronic CAD demonstrated an area under the curve of 0.722 (p < 0.001) with 79% sensitivity and 70% specificity. Using in silico gene analysis, we detected miR-483-5p cardiac gene targets, responsible for inflammation (PLA2G5), oxidative stress (NUDT8, GRK2), apoptosis (DNAAF10), fibrosis (IQSEC2, ZMYM6, MYOM2), angiogenesis (HGSNAT, TIMP2) and wound healing (ADAMTS2). High miR-483-5p 'coronary arterial gradient' in acute-MI, unnoticed in chronic-CAD, suggests important local mechanisms for miR483-5p in CAD in response to local myocardial ischemia. MiR-483-5p may have an important role as a gene modulator for pathologic and tissue repair states, is a suggestive biomarker, and is a potential therapeutic target for acute and chronic cardiovascular disease.
Collapse
Affiliation(s)
- Olga Volodko
- The Lydia and Carol Kittner, Lea and Benjamin Davidai Division of Cardiovascular Medicine and Surgery and Research Institute, Tzafon Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Tiberias 1528001, Israel
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Natalia Volinsky
- The Lydia and Carol Kittner, Lea and Benjamin Davidai Division of Cardiovascular Medicine and Surgery and Research Institute, Tzafon Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Tiberias 1528001, Israel
| | - Merav Yarkoni
- Heart Institute, Hadassah University Medical Center, Jerusalem, Department of Cardiology, Hadassah Medical Center, Faculty of Medicine, Hebrew University Jerusalem, Jerusalem 9574409, Israel
| | - Nufar Margalit
- The Lydia and Carol Kittner, Lea and Benjamin Davidai Division of Cardiovascular Medicine and Surgery and Research Institute, Tzafon Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Tiberias 1528001, Israel
| | - Fabio Kusniec
- The Lydia and Carol Kittner, Lea and Benjamin Davidai Division of Cardiovascular Medicine and Surgery and Research Institute, Tzafon Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Tiberias 1528001, Israel
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Doron Sudarsky
- The Lydia and Carol Kittner, Lea and Benjamin Davidai Division of Cardiovascular Medicine and Surgery and Research Institute, Tzafon Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Tiberias 1528001, Israel
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Gabby Elbaz-Greener
- Heart Institute, Hadassah University Medical Center, Jerusalem, Department of Cardiology, Hadassah Medical Center, Faculty of Medicine, Hebrew University Jerusalem, Jerusalem 9574409, Israel
| | - Shemy Carasso
- The Lydia and Carol Kittner, Lea and Benjamin Davidai Division of Cardiovascular Medicine and Surgery and Research Institute, Tzafon Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Tiberias 1528001, Israel
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Offer Amir
- The Lydia and Carol Kittner, Lea and Benjamin Davidai Division of Cardiovascular Medicine and Surgery and Research Institute, Tzafon Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Tiberias 1528001, Israel
- Heart Institute, Hadassah University Medical Center, Jerusalem, Department of Cardiology, Hadassah Medical Center, Faculty of Medicine, Hebrew University Jerusalem, Jerusalem 9574409, Israel
| |
Collapse
|
12
|
Buckels EJ, Hsu HL, Buchanan CM, Matthews BG, Lee KL. Genetic ablation of the preptin-coding portion of Igf2 impairs pancreatic function in female mice. Am J Physiol Endocrinol Metab 2022; 323:E467-E479. [PMID: 36459047 DOI: 10.1152/ajpendo.00401.2021] [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] [Indexed: 11/22/2022]
Abstract
Preptin is a 34-amino acid peptide derived from the E-peptide of pro-insulin-like growth factor 2 and is co-secreted with insulin from β-cells. Little is understood about the effects of endogenous preptin on whole body glucose metabolism. We developed a novel mouse model in which the preptin portion of Igf2 was genetically ablated in all tissues, hereafter referred to as preptin knockout (KO), and tested the hypothesis that the removal of preptin will lead to a decreased insulin response to a metabolic challenge. Preptin KO and wild-type (WT) mice underwent weekly fasting blood glucose measurements, intraperitoneal insulin tolerance tests (ITT) at 9, 29, and 44 wk of age, and an oral glucose tolerance test (GTT) at 45 wk of age. Preptin KO mice of both sexes had similar Igf2 exon 2-3 mRNA expression in the liver and kidney compared with WT mice, but Igf2 exon 3-4 (preptin) expression was not detectable. Western blot analysis of neonatal serum indicated that processing of pro-IGF2 translated from the KO allele may be altered. Preptin KO mice had similar body weight, body composition, β-cell area, and fasted glucose concentrations compared with WT mice in both sexes up to 47 wk of age. Female KO mice had a diminished ability to mount an insulin response following glucose stimulation in vivo. This effect was absent in male KO mice. Although preptin is not essential for glucose homeostasis, when combined with previous in vitro and ex vivo findings, these data show that preptin positively impacts β-cell function.NEW & NOTEWORTHY This is the first study to describe a model in which the preptin-coding portion of the Igf2 gene has been genetically ablated in mice. The mice do not show reduced size at birth associated with Igf2 knockout suggesting that IGF2 functionality is maintained, yet we demonstrate a change in the processing of mature Igf2. Female knockout mice have diminished glucose-stimulated insulin secretion, whereas the insulin response in males is not different to wild type.
Collapse
Affiliation(s)
- E J Buckels
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, New Zealand
| | - H-L Hsu
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand
| | - C M Buchanan
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, New Zealand
| | - B G Matthews
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, New Zealand
| | - K L Lee
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, New Zealand
| |
Collapse
|
13
|
Wang Z, Mohan R, Chen X, Matson K, Waugh J, Mao Y, Zhang S, Li W, Tang X, Satin LS, Tang X. microRNA-483 Protects Pancreatic β-Cells by Targeting ALDH1A3. Endocrinology 2021; 162:6132087. [PMID: 33564883 PMCID: PMC7951052 DOI: 10.1210/endocr/bqab031] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Indexed: 12/14/2022]
Abstract
Pancreatic β-cell dysfunction is central to the development and progression of type 2 diabetes. Dysregulation of microRNAs (miRNAs) has been associated with pancreatic islet dysfunction in type 2 diabetes. Previous study has shown that miR-483 is expressed relatively higher in β-cells than in α-cells. To explore the physiological function of miR-483, we generated a β-cell-specific knockout mouse model of miR-483. Loss of miR-483 enhances high-fat diet-induced hyperglycemia and glucose intolerance by the attenuation of diet-induced insulin release. Intriguingly, mice with miR-483 deletion exhibited loss of β-cell features, as indicated by elevated expression of aldehyde dehydrogenase family 1, subfamily A3 (Aldh1a3), a marker of β-cell dedifferentiation. Moreover, Aldh1a3 was validated as a direct target of miR-483 and overexpression of miR-483 repressed Aldh1a3 expression. Genetic ablation of miR-483 also induced alterations in blood lipid profile. Collectively, these data suggest that miR-483 is critical in protecting β-cell function by repressing the β-cell disallowed gene Aldh1a3. The dysregulated miR-483 may impair insulin secretion and initiate β-cell dedifferentiation during the development of type 2 diabetes.
Collapse
Affiliation(s)
- Zhihong Wang
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Ramkumar Mohan
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Xinqian Chen
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Katy Matson
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Jackson Waugh
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Yiping Mao
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Shungang Zhang
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Wanzhen Li
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Xiaohu Tang
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Leslie S Satin
- Department of Pharmacology, Brehm Center for Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Xiaoqing Tang
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
- Correspondence: Xiaoqing Tang, PhD, Michigan Technological University, 1400 Townsend Dr., Houghton, MI 49931, USA.
| |
Collapse
|
14
|
Sandovici I, Hammerle CM, Virtue S, Vivas-Garcia Y, Izquierdo-Lahuerta A, Ozanne SE, Vidal-Puig A, Medina-Gómez G, Constância M. Autocrine IGF2 programmes β-cell plasticity under conditions of increased metabolic demand. Sci Rep 2021; 11:7717. [PMID: 33833312 PMCID: PMC8032793 DOI: 10.1038/s41598-021-87292-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
When exposed to nutrient excess and insulin resistance, pancreatic β-cells undergo adaptive changes in order to maintain glucose homeostasis. The role that growth control genes, highly expressed in early pancreas development, might exert in programming β-cell plasticity in later life is a poorly studied area. The imprinted Igf2 (insulin-like growth factor 2) gene is highly transcribed during early life and has been identified in recent genome-wide association studies as a type 2 diabetes susceptibility gene in humans. Hence, here we investigate the long-term phenotypic metabolic consequences of conditional Igf2 deletion in pancreatic β-cells (Igf2βKO) in mice. We show that autocrine actions of IGF2 are not critical for β-cell development, or for the early post-natal wave of β-cell remodelling. Additionally, adult Igf2βKO mice maintain glucose homeostasis when fed a chow diet. However, pregnant Igf2βKO females become hyperglycemic and hyperinsulinemic, and their conceptuses exhibit hyperinsulinemia and placentomegalia. Insulin resistance induced by congenital leptin deficiency also renders Igf2βKO females more hyperglycaemic compared to leptin-deficient controls. Upon high-fat diet feeding, Igf2βKO females are less susceptible to develop insulin resistance. Based on these findings, we conclude that in female mice, autocrine actions of β-cell IGF2 during early development determine their adaptive capacity in adult life.
Collapse
Affiliation(s)
- Ionel Sandovici
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Department of Obstetrics and Gynaecology and National Institute for Health Research, Cambridge Biomedical Research Centre, Cambridge, CB2 0SW, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK.
| | - Constanze M Hammerle
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Department of Obstetrics and Gynaecology and National Institute for Health Research, Cambridge Biomedical Research Centre, Cambridge, CB2 0SW, UK.
- Novo Nordisk A/S, 2880, Bagsværd, Denmark.
| | - Sam Virtue
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Yurena Vivas-Garcia
- Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Madrid, Spain
- Nuffield Department of Clinical Medicine, Ludwig Institute for Cancer Research, University of Oxford, Headington, Oxford, OX3 7DQ, UK
| | - Adriana Izquierdo-Lahuerta
- Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Madrid, Spain
| | - Susan E Ozanne
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Antonio Vidal-Puig
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
- Welcome Trust Sanger Institute, Hinxton, CB10 1SA, UK
- Cambridge University Nanjing Centre of Technology and Innovation, Jiangbei Area, Nanjing, People's Republic of China
| | - Gema Medina-Gómez
- Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Madrid, Spain
| | - Miguel Constância
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Department of Obstetrics and Gynaecology and National Institute for Health Research, Cambridge Biomedical Research Centre, Cambridge, CB2 0SW, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK.
| |
Collapse
|
15
|
Foudi N, Legeay S. Effects of physical activity on cell-to-cell communication during type 2 diabetes: A focus on miRNA signaling. Fundam Clin Pharmacol 2021; 35:808-821. [PMID: 33675090 DOI: 10.1111/fcp.12665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 02/13/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes (TD2) is a progressive disease characterized by hyperglycemia that results from alteration in insulin secretion, insulin resistance, or both. A number of alterations involving different tissues and organs have been reported to the development and the progression of T2D, and more relevantly, through cell-to-cell communication pathways. Recent studies demonstrated that miRNAs are considerably implicated to cell-to-cell communication during T2D. Physical activity (PA) is associated with decreasing risks of developing T2D and acts as insulin-like factor. Cumulative evidence suggests that this effect could be mediated in part through improving insulin sensitivity in T2D and obese patients and modulating miRNAs synthesis and release in healthy patients. Therefore, the practice of PA should ideally be established before the initiation of T2D. This review describes cell-to-cell communications involved in the pathophysiology of T2D during PA.
Collapse
Affiliation(s)
- Nabil Foudi
- Department of Pharmacy, UNIV Angers, Angers, France.,Faculty of Medicine, Department of Pharmacy, University Ferhat Abbas Setif 1, Setif, Algeria
| | - Samuel Legeay
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, IRIS-IBS-CHU, Angers, France
| |
Collapse
|
16
|
He Q, Bo J, Shen R, Li Y, Zhang Y, Zhang J, Yang J, Liu Y. S1P Signaling Pathways in Pathogenesis of Type 2 Diabetes. J Diabetes Res 2021; 2021:1341750. [PMID: 34751249 PMCID: PMC8571914 DOI: 10.1155/2021/1341750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of type 2 diabetes mellitus (T2DM) is very complicated. The currently well-accepted etiology is the "Ominous Octet" theory proposed by Professor Defronzo. Since presently used drugs for T2DM have limitations and harmful side effects, studies regarding alternative treatments are being conducted. Analyzing the pharmacological mechanism of biomolecules in view of pathogenesis is an effective way to assess new drugs. Sphingosine 1 phosphate (S1P), an endogenous lipid substance in the human body, has attracted increasing attention in the T2DM research field. This article reviews recent study updates of S1P, summarizing its effects on T2DM with respect to pathogenesis, promoting β cell proliferation and inhibiting apoptosis, reducing insulin resistance, protecting the liver and pancreas from lipotoxic damage, improving intestinal incretin effects, lowering basal glucagon levels, etc. With increasing research, S1P may help treat and prevent T2DM in the future.
Collapse
Affiliation(s)
- Qiong He
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Jiaqi Bo
- Department of Second Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Ruihua Shen
- Department of Second Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Yan Li
- Department of Second Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Jiaxin Zhang
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Jing Yang
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| |
Collapse
|
17
|
Li S, Li H, Ge W, Song K, Yuan C, Yin R. Effect of miR-184 on Proliferation and Apoptosis of Pancreatic Ductal Adenocarcinoma and Its Mechanism. Technol Cancer Res Treat 2020; 19:1533033820943237. [PMID: 32914707 PMCID: PMC7488881 DOI: 10.1177/1533033820943237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Objective: Previous studies have shown that abnormal expression of microRNA-184 leads to a variety of cancers, including pancreatic ductal adenocarcinoma, suggesting microRNA-184 as a new treatment target for pancreatic ductal adenocarcinoma. However, the molecular mechanism of microRNA-184 in pancreatic ductal adenocarcinoma remains unclear. It is important to investigate the effect and role of microRNA-184 in pancreatic ductal adenocarcinoma. Methods: The clinical and laboratory inspection data of 120 patients with pancreatic cancer admitted to the First Affiliated Hospital of Anhui Medical University were compared. MicroRNA-184 expression in tumor tissues and cells was evaluated using reverse transcription polymerase chain reaction. Flow cytometry and Annexin V/propidium iodide staining were performed to examine cell cycle and apoptosis. Western blotting analysis was conducted to measure the protein expression of p-PI3K, p-AKT, JNK1, C-Myc, C-Jun, caspase-9, and caspase-3. Results: MicroRNA-184 expression was low in patients with pancreatic ductal adenocarcinoma. Survival curve showed that patients with lower expression of microRNA-184 in tumor tissues had a worse prognosis and shorter survival time (P < .05), and the multivariate analysis identified that microRNA-184 was an independent prognostic indicator (P < .05). In vitro studies showed that microRNA-184 overexpression induced apoptosis and suppressed cell cycle transition from G1 to S and G2 phases in pancreatic ductal adenocarcinoma cells. Furthermore, molecular studies revealed that inhibition of microRNA-184 promoted the gene expression of p-PI3K, p-AKT, JNK1, C-Myc, and C-Jun compared with the control group. Overexpression of microRNA-184 led to significantly increased expression of caspase-9 and caspase-3 and significantly decreased expression of Bcl-2. Conclusion: This study suggests that microRNA-184 inhibits the proliferation and promotes the apoptosis of pancreatic ductal adenocarcinoma cells by downregulating the expression of C-Myc, C-Jun, and Bcl-2. Our verification of the role of microRNA-184 may provide a novel biomarker for the diagnosis, therapy, and prognosis of pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Shentao Li
- Department of Emergency, 533251Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - He Li
- Department of Emergency, 533251Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Weiwei Ge
- Department of Emergency, 533251Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Kai Song
- Department of Emergency, 533251Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chunyu Yuan
- Department of Emergency, 533251Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ran Yin
- Department of Emergency, 533251Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
18
|
Kaur P, Kotru S, Singh S, Behera BS, Munshi A. Role of miRNAs in the pathogenesis of T2DM, insulin secretion, insulin resistance, and β cell dysfunction: the story so far. J Physiol Biochem 2020; 76:485-502. [PMID: 32749641 DOI: 10.1007/s13105-020-00760-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 07/29/2020] [Indexed: 01/24/2023]
Abstract
Diabetes, the most common endocrine disorder, also known as a silent killer disease, is characterized by uncontrolled hyperglycemia. According to the International Diabetes Federation, there were 451 million people with diabetes mellitus worldwide in 2017. It is a multifactorial syndrome caused by genetic as well as environmental factors. Noncoding RNAs, especially the miRNAs, play a significant role in the development as well as the progression of the disease. This is on account of insulin resistance or defects in β cell function. Various miRNAs including miR-7, miR-9, miR-16, miR-27, miR-24, miR-29, miR-124a, miR-135, miR-130a, miR-144, miR-181a, and miR-375 and many more have been associated with insulin resistance and other pathogenic conditions leading to the development of the disease. These miRNAs play significant roles in various pathways underlying insulin resistance such as PI3K, AKT/GSK, and mTOR. The main target genes of these miRNAs are FOXO1, FOXA2, STAT3, and PTEN. The miRNAs carry out important functions in insulin target tissues like the adipose tissue, liver, and muscle. MiRNAs miR-9, miR-375, and miR-124a, are also associated with the secretion of insulin from pancreatic cells. There is an interplay between the miRNAs and pancreatic cell growth, especially the miRNAs affecting development and proliferation of these cells. Most of the miRNAs target more than one gene which not only justifies their use as biomarkers but also their therapeutic potential. The current review has been compiled with an aim to discuss the role of various miRNAs involved in various pathogenic mechanisms including insulin resistance, insulin secretion, and the β cell dysfunction.
Collapse
Affiliation(s)
- Prabhsimran Kaur
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151001, India
| | - Sushil Kotru
- Max Endocrinology, Diabetes and Obesity Care Centre, Max Superspeciality Hospital, Bathinda, 151001, India
| | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151001, India
| | - Bidwan Sekhar Behera
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151001, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151001, India.
| |
Collapse
|
19
|
Jiang Y, Rosborough BR, Chen J, Das S, Kitsios GD, McVerry BJ, Mallampalli RK, Lee JS, Ray A, Chen W, Ray P. Single cell RNA sequencing identifies an early monocyte gene signature in acute respiratory distress syndrome. JCI Insight 2020; 5:135678. [PMID: 32554932 PMCID: PMC7406263 DOI: 10.1172/jci.insight.135678] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 06/03/2020] [Indexed: 01/16/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) results from overwhelming pulmonary inflammation. Prior bulk RNA sequencing provided limited insights into ARDS pathogenesis. We used single cell RNA sequencing to probe ARDS at a higher resolution. PBMCs of patients with pneumonia and sepsis with early ARDS were compared with those of sepsis patients who did not develop ARDS. Monocyte clusters from ARDS patients revealed multiple distinguishing characteristics in comparison with monocytes from patients without ARDS, including downregulation of SOCS3 expression, accompanied by a proinflammatory signature with upregulation of multiple type I IFN-induced genes, especially in CD16+ cells. To generate an ARDS risk score, we identified upregulation of 29 genes in the monocytes of these patients, and 17 showed a similar profile in cells of patients in independent cohorts. Monocytes had increased expression of RAB11A, known to inhibit neutrophil efferocytosis; ATP2B1, a calcium pump that exports Ca2+ implicated in endothelial barrier disruption; and SPARC, associated with processing of procollagen to collagen. These data show that monocytes of ARDS patients upregulate expression of genes not just restricted to those associated with inflammation. Together, our findings identify molecules that are likely involved in ARDS pathogenesis that may inform biomarker and therapeutic development.
Collapse
Affiliation(s)
- Yale Jiang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,School of Medicine, Tsinghua University, Beijing, China
| | - Brian R. Rosborough
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Jie Chen
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Sudipta Das
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Georgios D. Kitsios
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Bryan J. McVerry
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Rama K. Mallampalli
- Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart Lung Research Institute, Columbus, Ohio
| | - Janet S. Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Anuradha Ray
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Wei Chen
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Prabir Ray
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
20
|
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: 1.8] [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
|
21
|
Zheng H, Li X, Yang X, Yan F, Wang C, Liu J. miR-217/Mafb Axis Involve in High Glucose-Induced β-TC-tet Cell Damage Via Regulating NF-κB Signaling Pathway. Biochem Genet 2020; 58:901-913. [PMID: 32627107 DOI: 10.1007/s10528-020-09984-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 06/25/2020] [Indexed: 12/12/2022]
Abstract
We attempt to explore the role of miR-217 during the process of type 2 diabetes mellitus (T2DM). Mouse β-TC-tet was dealt with 16.7 mM glucose (HG) to imitate the cells in T2DM. Cell proliferation and apoptosis were determined by cell counting kit-8 and flow cytometry. The correlation between miR-217 and Mafb was predicted with biological software and confirmed by dual lucifierase assay. Western blot was applied to detect protein expression. The data from GEO database exhibited that miR-217 was upregulated in T2DM patients. HG treatment upregulated the expression of miR-217, inhibited the proliferation, and promoted the apoptosis and inflammation of β-TC-tet cell. Depletion of miR-217 alleviated the damage of β-TC-tet cell caused by HG. Mafb was affirmed as a target of miR-217 and was negatively modulated by miR-217. Knockdown of Mafb attenuated the alleviation of miR-217 inhibitor on β-TC-tet cell damage. The expression of key proteins in NF-κB signaling pathway was upregulated by HG, and this upregulation tendency was inhibited by miR-217 inhibitor. Moreover, silencing Mafb could alleviate the inhibition of miR-217 inhibitor on these proteins. Our findings insinuated that inhibition of miR-217 could relieve β-TC-tet damage induced by HG through regulating Mafb and NF-κB signaling.
Collapse
Affiliation(s)
- Huizhen Zheng
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, P.R. China.,Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, 250012, Shandong, P.R. China.,Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, 250012, Shandong, P.R. China.,Jinan Clinical Research Center for Endocrine and Metabolic Diseases, Jinan, 250012, Shandong, P.R. China
| | - Xinying Li
- Department of Emergency, Shandong Provincial Western Hospital, Shandong Provincial ENT Hospital, Jinan, Shandong, P.R. China
| | - Xinyu Yang
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Fei Yan
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, P.R. China.,Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, 250012, Shandong, P.R. China.,Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, 250012, Shandong, P.R. China.,Jinan Clinical Research Center for Endocrine and Metabolic Diseases, Jinan, 250012, Shandong, P.R. China
| | - Chuan Wang
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, P.R. China.,Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, 250012, Shandong, P.R. China.,Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, 250012, Shandong, P.R. China.,Jinan Clinical Research Center for Endocrine and Metabolic Diseases, Jinan, 250012, Shandong, P.R. China
| | - Jinbo Liu
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, P.R. China. .,Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, 250012, Shandong, P.R. China. .,Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, 250012, Shandong, P.R. China. .,Jinan Clinical Research Center for Endocrine and Metabolic Diseases, Jinan, 250012, Shandong, P.R. China.
| |
Collapse
|
22
|
Effect of imbalance in folate and vitamin B12 in maternal/parental diet on global methylation and regulatory miRNAs. Sci Rep 2019; 9:17602. [PMID: 31772242 PMCID: PMC6879517 DOI: 10.1038/s41598-019-54070-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
DNA methylation, a central component of the epigenetic network is altered in response to nutritional influences. In one-carbon cycle, folate acts as a one-carbon carrier and vitamin B12 acts as co-factor for the enzyme methionine synthase. Both folate and vitamin B12 are the important regulators of DNA methylation which play an important role in development in early life. Previous studies carried out in this regard have shown the individual effects of these vitamins but recently the focus has been to study the combined effects of both the vitamins during pregnancy. Therefore, this study was planned to elucidate the effect of the altered dietary ratio of folate and B12 on the expression of transporters, related miRNAs and DNA methylation in C57BL/6 mice. Female mice were fed diets with 9 combinations of folate and B12 for 4 weeks. They were mated and off-springs born (F1) were continued on the same diet for 6 weeks post-weaning. Maternal and fetal (F2) tissues were collected at day 20 of gestation. Deficient state of folate led to an increase in the expression of folate transporters in both F1 and F2 generations, however, B12 deficiency (BDFN) also led to an increase in the expression in both the generations. B12 transporters/proteins were found to be increased with B12 deficiency in F1 and F2 generations except for TC-II in the kidney which was found to be decreased in the F1 generation. miR-483 was found to be increased with all conditions of folate and B12 in both F1 and F2 generations, however, deficient conditions of B12 led to an increase in the expression of miR-221 in both F1 and F2 generations. The level of miR-133 was found to be increased in BDFN group in F1 generation however; in F2 generation the change in expression was tissue and sex-specific. Global DNA methylation was decreased with deficiency of both folate and B12 in maternal tissues (F1) but increased with folate deficiency in placenta (F1) and under all conditions in fetal tissues (F2). DNA methyltransferases were overall found to be increased with deficiency of folate and B12 in both F1 and F2 generations. Results suggest that the dietary ratio of folate and B12 resulted in altered expression of transporters, miRNAs, and genomic DNA methylation in association with DNMTs.
Collapse
|
23
|
Hao Y, Wang W, Wu D, Liu K, Sun Y. Retracted: Bilobalide alleviates tumor necrosis factor‐alpha‐induced pancreatic beta‐cell MIN6 apoptosis and dysfunction through upregulation of miR‐153. Phytother Res 2019; 34:409-417. [PMID: 31667906 DOI: 10.1002/ptr.6533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/22/2019] [Accepted: 10/09/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Yan Hao
- Department of EndocrinologyJining No.1 People's Hospital Jining China
| | - Weiwei Wang
- Department of EndocrinologyJining No.1 People's Hospital Jining China
| | - Dong Wu
- Emergency DepartmentJining No.1 People's Hospital Jining China
| | - Kai Liu
- Emergency DepartmentJinxiang People's Hospital Jining China
| | - Yihan Sun
- Department of EndocrinologyJining No.1 People's Hospital Jining China
| |
Collapse
|
24
|
Holly JMP, Biernacka K, Perks CM. The Neglected Insulin: IGF-II, a Metabolic Regulator with Implications for Diabetes, Obesity, and Cancer. Cells 2019; 8:cells8101207. [PMID: 31590432 PMCID: PMC6829378 DOI: 10.3390/cells8101207] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023] Open
Abstract
When originally discovered, one of the initial observations was that, when all of the insulin peptide was depleted from serum, the vast majority of the insulin activity remained and this was due to a single additional peptide, IGF-II. The IGF-II gene is adjacent to the insulin gene, which is a result of gene duplication, but has evolved to be considerably more complicated. It was one of the first genes recognised to be imprinted and expressed in a parent-of-origin specific manner. The gene codes for IGF-II mRNA, but, in addition, also codes for antisense RNA, long non-coding RNA, and several micro RNA. Recent evidence suggests that each of these have important independent roles in metabolic regulation. It has also become clear that an alternatively spliced form of the insulin receptor may be the principle IGF-II receptor. These recent discoveries have important implications for metabolic disorders and also for cancer, for which there is renewed acknowledgement of the importance of metabolic reprogramming.
Collapse
Affiliation(s)
- Jeff M P Holly
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK.
| | - Kalina Biernacka
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Claire M Perks
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| |
Collapse
|
25
|
Ginés I, Gil-Cardoso K, Serrano J, Casanova-Marti À, Lobato M, Terra X, Blay MT, Ardévol A, Pinent M. Proanthocyanidins Limit Adipose Accrual Induced by a Cafeteria Diet, Several Weeks after the End of the Treatment. Genes (Basel) 2019; 10:genes10080598. [PMID: 31398921 PMCID: PMC6723337 DOI: 10.3390/genes10080598] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/19/2019] [Accepted: 08/05/2019] [Indexed: 11/25/2022] Open
Abstract
A dose of proanthocyanidins with satiating properties proved to be able to limit body weight increase several weeks after administration under exposure to a cafeteria diet. Here we describe some of the molecular targets and the duration of the effects. We treated rats with 500 mg grape seed proanthocyanidin extract (GSPE)/kg BW for ten days. Seven or seventeen weeks after the last GSPE dose, while animals were on a cafeteria diet, we used reverse transcriptase-polymerase chain reaction (RT-PCR) to measure the mRNA of the key energy metabolism enzymes from the liver, adipose depots and muscle. We found that a reduction in the expression of adipose Lpl might explain the lower amount of adipose tissue in rats seven weeks after the last GSPE dose. The liver showed increased expression of Cpt1a and Hmgs2 together with a reduction in Fasn and Dgat2. In addition, muscle showed a higher fatty oxidation (Oxct1 and Cpt1b mRNA). However, after seventeen weeks, there was a completely different gene expression pattern. At the conclusion of the study, seven weeks after the last GSPE administration there was a limitation in adipose accrual that might be mediated by an inhibition of the gene expression of the adipose tissue Lpl. Concomitantly there was an increase in fatty acid oxidation in liver and muscle.
Collapse
Affiliation(s)
- Iris Ginés
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
| | - Katherine Gil-Cardoso
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
| | - Joan Serrano
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
| | - Àngela Casanova-Marti
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
| | - Maria Lobato
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
| | - Ximena Terra
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
| | - M Teresa Blay
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
| | - Anna Ardévol
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain.
| | - Montserrat Pinent
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
| |
Collapse
|
26
|
Chen L, Pan X, Zhang YH, Huang T, Cai YD. Analysis of Gene Expression Differences between Different Pancreatic Cells. ACS OMEGA 2019; 4:6421-6435. [DOI: 10.1021/acsomega.8b02171] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Lei Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, China
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
- Shanghai Key Laboratory of PMMP, East China Normal University, Shanghai 200241, China
| | - Xiaoyong Pan
- Department of Medical Informatics, Erasmus MC, Rotterdam 3014ZK, Netherlands
| | - Yu-Hang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|
27
|
Nunez Lopez YO, Retnakaran R, Zinman B, Pratley RE, Seyhan AA. Predicting and understanding the response to short-term intensive insulin therapy in people with early type 2 diabetes. Mol Metab 2019; 20:63-78. [PMID: 30503831 PMCID: PMC6358589 DOI: 10.1016/j.molmet.2018.11.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/05/2018] [Accepted: 11/12/2018] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Short-term intensive insulin therapy (IIT) early in the course of type 2 diabetes acutely improves beta-cell function with long-lasting effects on glycemic control. However, conventional measures cannot determine which patients are better suited for IIT, and little is known about the molecular mechanisms determining response. Therefore, this study aimed to develop a model that could accurately predict the response to IIT and provide insight into molecular mechanisms driving such response in humans. METHODS Twenty-four patients with early type 2 diabetes were assessed at baseline and four weeks after IIT, consisting of basal detemir and premeal insulin aspart. Twelve individuals had a beneficial beta-cell response to IIT (responders) and 12 did not (nonresponders). Beta-cell function was assessed by multiple methods, including Insulin Secretion-Sensitivity Index-2. MicroRNAs (miRNAs) were profiled in plasma samples before and after IIT. The response to IIT was modeled using a machine learning algorithm and potential miRNA-mediated regulatory mechanisms assessed by differential expression, correlation, and functional network analyses (FNA). RESULTS Baseline levels of circulating miR-145-5p, miR-29c-3p, and HbA1c accurately (91.7%) predicted the response to IIT (OR = 121 [95% CI: 6.7, 2188.3]). Mechanistically, a previously described regulatory loop between miR-145-5p and miR-483-3p/5p, which controls TP53-mediated apoptosis, appears to also occur in our study population of humans with early type 2 diabetes. In addition, significant (fold change > 2, P < 0.05) longitudinal changes due to IIT in the circulating levels of miR-138-5p, miR-192-5p, miR-195-5p, miR-320b, and let-7a-5p further characterized the responder group and significantly correlated (|r| > 0.4, P < 0.05) with the changes in measures of beta-cell function and insulin sensitivity. FNA identified a network of coordinately/cooperatively regulated miRNA-targeted genes that potentially drives the IIT response through negative regulation of apoptotic processes that underlie beta cell dysfunction and concomitant positive regulation of proliferation. CONCLUSIONS Responses to IIT in people with early type 2 diabetes are associated with characteristic miRNA signatures. This study represents a first step to identify potential responders to IIT (a current limitation in the field) and provides important insight into the pathophysiologic determinants of the reversibility of beta-cell dysfunction. ClinicalTrial.gov identifier: NCT01270789.
Collapse
Affiliation(s)
- Yury O Nunez Lopez
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL 32804, USA
| | - Ravi Retnakaran
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Bernard Zinman
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Richard E Pratley
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL 32804, USA.
| | - Attila A Seyhan
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL 32804, USA; The Chemical Engineering Department, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
28
|
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: 3.2] [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
|
29
|
Zhao C, Yang C, Wai STC, Zhang Y, P. Portillo M, Paoli P, Wu Y, San Cheang W, Liu B, Carpéné C, Xiao J, Cao H. Regulation of glucose metabolism by bioactive phytochemicals for the management of type 2 diabetes mellitus. Crit Rev Food Sci Nutr 2018; 59:830-847. [PMID: 30501400 DOI: 10.1080/10408398.2018.1501658] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Chao Zhao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Department of Chemistry, University of California, Davis, CA, USA
| | - Chengfeng Yang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Institute of Oceanography, Minjiang University, Fuzhou, China
| | - Sydney Tang Chi Wai
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Yanbo Zhang
- School Chinese Medicine, University of Hong Kong, Hong Kong, China
| | - Maria P. Portillo
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Basque Country (UPV/EHU) and Lucio Lascaray Research Center, Vitoria, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), Spain
| | - Paolo Paoli
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Yijing Wu
- Institute of Oceanography, Minjiang University, Fuzhou, China
- College of Food Science and Nutritional Engineering, China Agricultural University, China
| | - Wai San Cheang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Control in Chinese Medicine, University of Macau, Macau SAR, China
| | - Bin Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Christian Carpéné
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM U1048)/Université Paul Sabatier, Bât. L4, CHU Rangueil, Toulouse cedex 4, France
| | - Jianbo Xiao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Control in Chinese Medicine, University of Macau, Macau SAR, China
| | - Hui Cao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Control in Chinese Medicine, University of Macau, Macau SAR, China
| |
Collapse
|
30
|
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: 41] [Impact Index Per Article: 6.8] [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
|
31
|
Abstract
Islets of Langerhans are islands of endocrine cells scattered throughout the pancreas. A number of new studies have pointed to the potential for conversion of non-β islet cells in to insulin-producing β-cells to replenish β-cell mass as a means to treat diabetes. Understanding normal islet cell mass and function is important to help advance such treatment modalities: what should be the target islet/β-cell mass, does islet architecture matter to energy homeostasis, and what may happen if we lose a particular population of islet cells in favour of β-cells? These are all questions to which we will need answers for islet replacement therapy by transdifferentiation of non-β islet cells to be a reality in humans. We know a fair amount about the biology of β-cells but not quite as much about the other islet cell types. Until recently, we have not had a good grasp of islet mass and distribution in the human pancreas. In this review, we will look at current data on islet cells, focussing more on non-β cells, and on human pancreatic islet mass and distribution.
Collapse
Affiliation(s)
- Gabriela Da Silva Xavier
- Section of Functional Genomics and Cell Biology, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK.
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Edgbaston B15 2TT, UK.
| |
Collapse
|
32
|
Mirra P, Nigro C, Prevenzano I, Leone A, Raciti GA, Formisano P, Beguinot F, Miele C. The Destiny of Glucose from a MicroRNA Perspective. Front Endocrinol (Lausanne) 2018; 9:46. [PMID: 29535681 PMCID: PMC5834423 DOI: 10.3389/fendo.2018.00046] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glucose serves as a primary, and for some tissues the unique, fuel source in order to generate and maintain the biological functions. Hyperglycemia is a hallmark of type 2 diabetes and is the direct consequence of perturbations in the glucose homeostasis. Insulin resistance, referred to as a reduced response of target tissues to the hormone, contributes to the development of hyperglycemia. The molecular mechanisms responsible for the altered glucose homeostasis are numerous and not completely understood. MicroRNAs (miRNAs) are now recognized as regulators of the lipid and glucose metabolism and are involved in the onset of metabolic diseases. Indeed, these small non-coding RNA molecules operate in the RNA silencing and posttranscriptional regulation of gene expression and may modulate the levels of kinases and enzymes in the glucose metabolism. Therefore, a better characterization of the function of miRNAs and a deeper understanding of their role in disease may represent a fundamental step toward innovative treatments addressing the causes, not only the symptoms, of hyperglycemia, using approaches aimed at restoring either miRNAs or their specific targets. In this review, we outline the current understanding regarding the impact of miRNAs in the glucose metabolism and highlight the need for further research focused on altered key kinases and enzymes in metabolic diseases.
Collapse
Affiliation(s)
- Paola Mirra
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Cecilia Nigro
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Immacolata Prevenzano
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Alessia Leone
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Gregory Alexander Raciti
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Pietro Formisano
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Francesco Beguinot
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Claudia Miele
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- *Correspondence: Claudia Miele,
| |
Collapse
|
33
|
LaPierre MP, Stoffel M. MicroRNAs as stress regulators in pancreatic beta cells and diabetes. Mol Metab 2017; 6:1010-1023. [PMID: 28951825 PMCID: PMC5605735 DOI: 10.1016/j.molmet.2017.06.020] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/29/2017] [Accepted: 06/02/2017] [Indexed: 12/12/2022] Open
Abstract
Background MicroRNAs have emerged as important regulatory non-coding RNAs that tune cellular responses to physiological perturbations and disease conditions. An increasing body of literature underlines the important roles of miRNA function in pancreatic β-cells in response to metabolic, genetic and inflammatory stress. Lessons from genetic loss- and gain-of-function studies have implicated several highly expressed and evolutionary conserved miRNAs in stress signal modulation, resolution and buffering, thereby forming stabilizing miRNA networks that preserve β-cell differentiation, function, proliferation and cell survival. Scope of Review This review will summarize our current knowledge of how biologically relevant miRNAs regulate stress responses in pancreatic β-cells, discuss the challenges and opportunities associated with using secreted miRNAs as biomarkers and forecast how mechanistic knowledge of miRNA function can be exploited in developing miRNA-based therapeutics. Major Conclusions miRNAs play important roles in the function, differentiation, proliferation, and survival of pancreatic β-cells. Many miRNA families that are regulated by metabolic, genetic, and inflammatory stressors have been found to coordinate the adaptive responses of β-cells in vivo in conditions such as obesity and diabetes.
Collapse
Affiliation(s)
| | - Markus Stoffel
- Corresponding author. Swiss Federal Institute of Technology, ETH Zürich, Institute for Molecular Health Science, Laboratory for Metabolic Diseases, Otto-Stern Weg 7, HPL H36, CH 8093 Zürich, Switzerland. Fax: +41 44 633 1362.Federal Institute of TechnologyETH ZürichInstitute for Molecular Health ScienceLaboratory for Metabolic DiseasesOtto-Stern Weg 7HPL H36ZürichCH 8093Switzerland
| |
Collapse
|
34
|
Wang H, Zhang H, Sun Q, Wang Y, Yang J, Yang J, Zhang T, Luo S, Wang L, Jiang Y, Zeng C, Cai D, Bai X. Intra-articular Delivery of Antago-miR-483-5p Inhibits Osteoarthritis by Modulating Matrilin 3 and Tissue Inhibitor of Metalloproteinase 2. Mol Ther 2017; 25:715-727. [PMID: 28139355 DOI: 10.1016/j.ymthe.2016.12.020] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/12/2016] [Accepted: 12/25/2016] [Indexed: 01/21/2023] Open
Abstract
MicroRNAs (miRNAs) are emerging as important regulators in osteoarthritis (OA) pathogenesis. In our study, a real-time PCR assay revealed that miR-483-5p was upregulated in articular cartilage from OA patients and experimental OA mice induced by destabilization of the medial meniscus compared to their controls. Overexpression of miR-483-5p by intra-articular injection of lentivirus LV3-miR-483-5p significantly enhanced the severity of experimental OA. Consequently, we synthesized antago-miR-483-5p to silence the endogenous miR-483-5p and delivered it intra-articularly, which revealed that antago-miR-483-5p delayed the progression of experimental OA. To investigate the functional mechanism of miR-483-5p in OA development, we generated doxycycline-inducible miR-483 transgenic (TG483) mice. TG483 mice exhibited significant acceleration and increased severity of OA, and age-related OA occurred with higher incidence and greater severity in TG483 mice compared with their controls. Furthermore, our results revealed miR-483-5p directly targeted to the cartilage matrix protein matrilin 3 (Matn3) and tissue inhibitor of metalloproteinase 2 (Timp2) to stimulate chondrocyte hypertrophy, extracellular matrix degradation, and cartilage angiogenesis, and it consequently initiated and accelerated the development of OA. In conclusion, our findings reveal an miRNA functional pathway important for OA development. Targeting of miR-483-5p by intra-articular injection of antago-miR-483-5p represents an approach that could prevent the onset of OA and delay its progression.
Collapse
Affiliation(s)
- Hua Wang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; Key Laboratory of Tropical Diseases and Translational Medicine of The Ministry of Education, Hainan Medical College, Haikou 571199, China
| | - Haiyan Zhang
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Qiuyi Sun
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Yun Wang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Jun Yang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Jincheng Yang
- Department of Orthopedics, General Hospital of Guangzhou Military Command of PLA, Guangzhou 510010, China
| | - Tao Zhang
- Department of Orthopedics, General Hospital of Guangzhou Military Command of PLA, Guangzhou 510010, China
| | - Shenqiu Luo
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Liping Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chun Zeng
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China.
| | - Daozhang Cai
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China.
| | - Xiaochun Bai
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China.
| |
Collapse
|
35
|
Hu S, Zhang M, Sun F, Ren L, He X, Hua J, Peng S. miR-375 controls porcine pancreatic stem cell fate by targeting 3-phosphoinositide-dependent protein kinase-1 (Pdk1). Cell Prolif 2017; 49:395-406. [PMID: 27218665 DOI: 10.1111/cpr.12263] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/10/2016] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES miR-375 is one of the highly expressed microRNAs (miRNAs) found in pancreatic islets of both humans and mice. In this study, we investigated functions of miRNA miR-375 in porcine pancreatic stem cells (PSC). MATERIALS AND METHODS We transfected mimic and inhibitor of miR-375 in PSCs to measure functional roles of the microRNA and its effects on cell cycle proliferation and cell differentiation were determined. Luciferase assays were also performed to reveal the target gene of miR-375. RESULTS Overexpression of miR-375 suppressed proliferation, promoted apoptosis and inhibited differentiation into islet-like cells. PDK1 was identified as being a target of miR-375. Furthermore, we found that overexpression of miR-375 inhibited activation of the PDK1-AKT signalling pathway. CONCLUSION miR-375 directly targeted PDK1 in porcine PSCs, suppressing cell proliferation and differentiation into islet-like cells.
Collapse
Affiliation(s)
- Shuxian Hu
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, Shaanxi, China
| | - Mingzhi Zhang
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, Shaanxi, China
| | - Fen Sun
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, Shaanxi, China
| | - Lipeng Ren
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, Shaanxi, China
| | - Xin He
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, Shaanxi, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, Shaanxi, China
| | - Sha Peng
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
36
|
Dalgaard LT, Eliasson L. An 'alpha-beta' of pancreatic islet microribonucleotides. Int J Biochem Cell Biol 2017; 88:208-219. [PMID: 28122254 DOI: 10.1016/j.biocel.2017.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 11/17/2022]
Abstract
MicroRNAs (miRNAs) are cellular, short, non-coding ribonucleotides acting as endogenous posttranscriptional repressors following incorporation in the RNA-induced silencing complex. Despite being chemically and mechanistically very similar, miRNAs exert a multitude of different cellular effects by acting on mRNA species, whose gene-products partake in a wide array of processes. Here, the aim was to review the knowledge of miRNA expression and action in the islet of Langerhans. We have focused on: 1) physiological consequences of islet or beta cell specific inhibition of miRNA processing, 2) mechanisms regulating processing of miRNAs in islet cells, 3) presence and function of miRNAs in alpha versus beta cells - the two main cell types of islets, and 4) miRNA mediators of beta cell decompensation. It is clear that miRNAs regulate pancreatic islet development, maturation, and function in vivo. Moreover, processing of miRNAs appears to be altered by obesity, diabetes, and aging. A number of miRNAs (such as miR-7, miR-21, miR-29, miR-34a, miR-212/miR-132, miR-184, miR-200 and miR-375) are involved in mediating beta cell dysfunction and/or compensation induced by hyperglycemia, oxidative stress, cytotoxic cytokines, and in rodent models of fetal metabolic programming prediabetes and overt diabetes. Studies of human type 2 diabetic islets underline that these miRNA families could have important roles also in human type 2 diabetes. Furthermore, there is a genuine gap of knowledge regarding miRNA expression and function in pancreatic alpha cells. Progress in this area would be enhanced by improved in vitro alpha cell models and better tools for islet cell sorting.
Collapse
Affiliation(s)
| | - Lena Eliasson
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, CRC, SUS, Malmö, Sweden.
| |
Collapse
|
37
|
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: 11.6] [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
|
38
|
Calderari S, Diawara MR, Garaud A, Gauguier D. Biological roles of microRNAs in the control of insulin secretion and action. Physiol Genomics 2016; 49:1-10. [PMID: 27815534 DOI: 10.1152/physiolgenomics.00079.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/02/2016] [Indexed: 02/03/2023] Open
Abstract
microRNAs (miRNAs) are intracellular and circulating molecular components contributing to genome expression control through binding mRNA targets, which generally results in downregulated mRNA expression. One miRNA can target several mRNAs, and one transcript can be targeted by several miRNAs, resulting in complex fine-tuning of regulation of gene networks and signaling pathways. miRNAs regulate metabolism, adipocyte differentiation, pancreatic development, β-cell mass, insulin biosynthesis, secretion, and signaling, and their role in diabetes and obesity is emerging. Their pathophysiological effects are essentially dependent on cellular coexpression with their mRNA targets, which can show tissue-specific transcriptional responses to disease conditions and environmental challenges. Current knowledge of miRNA biology and their impact on the pathogenesis of diabetes and obesity is based on experimental data documenting miRNA expression generally in single tissue types that can be correlated with expression of target mRNAs to integrate miRNA in functional pathways and gene networks. Here we present results from the most significant studies dealing with miRNA function in liver, fat, skeletal muscle, and endocrine pancreas and their implication in diabetes and obesity.
Collapse
Affiliation(s)
- Sophie Calderari
- Sorbonne Universities, University Pierre & Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM UMR_S 1138, Cordeliers Research Centre, Paris, France; and.,Institut National de la Recherche Agronomique, ENVA, University Paris Saclay, Jouy en Josas, France
| | - Malika R Diawara
- Sorbonne Universities, University Pierre & Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM UMR_S 1138, Cordeliers Research Centre, Paris, France; and
| | - Alois Garaud
- Sorbonne Universities, University Pierre & Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM UMR_S 1138, Cordeliers Research Centre, Paris, France; and
| | - Dominique Gauguier
- Sorbonne Universities, University Pierre & Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM UMR_S 1138, Cordeliers Research Centre, Paris, France; and
| |
Collapse
|
39
|
Ma H, Wu Y, Yang H, Liu J, Dan H, Zeng X, Zhou Y, Jiang L, Chen Q. MicroRNAs in oral lichen planus and potential miRNA-mRNA pathogenesis with essential cytokines: a review. Oral Surg Oral Med Oral Pathol Oral Radiol 2016; 122:164-73. [PMID: 27282956 DOI: 10.1016/j.oooo.2016.03.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 01/20/2016] [Accepted: 03/17/2016] [Indexed: 02/05/2023]
Abstract
Oral lichen planus (OLP) is a potentially premalignant condition with unknown pathogenesis. Immune and inflammatory factors are thought to play important roles in the development of OLP, and cytokines, such as interferon (IFN)-γ and tumor necrosis factor (TNF)-α, can act as critical players in the immunopathogenesis of OLP. MicroRNAs (miRNAs) are closely correlated with cytokines in various inflammation-related diseases. In patients with OLP, miRNA-146a and miRNA-155 are increased in peripheral blood mononuclear cells, and numerous miRNAs have been shown to exhibit altered expression profiles in lesions. Although the microRNA-messenger RNA (miRNA-mRNA) network is thought to be involved in the development of OLP, in-depth studies are lacking. Here, we summarize current data on the mechanisms of action of miRNAs regulating typical cytokines in OLP, including interleukin (IL)-10, IL-17, IL-22, IFN-γ, and TNF-α, to study the genetic basis of the pathogenesis of OLP and to provide prospects of therapy.
Collapse
Affiliation(s)
- Hui Ma
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuanqin Wu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Huamei Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiajia Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hongxia Dan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Lu Jiang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|