1
|
Shnayder NA, Pekarets NA, Pekarets NI, Dmitrenko DV, Grechkina VV, Petrova MM, Al-Zamil M, Nasyrova RF. MicroRNAs as Epigenetic Biomarkers of Pathogenetic Mechanisms of the Metabolic Syndrome Induced by Antiseizure Medications: Systematic Review. J Clin Med 2025; 14:2432. [PMID: 40217882 PMCID: PMC11989458 DOI: 10.3390/jcm14072432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/27/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Antiseizure medication (ASM) induced metabolic syndrome (AIMetS) is a common adverse drug reaction (ADR) of pharmacotherapy for epilepsy and psychiatric disorders. However, the sensitivity and specificity of blood biomarkers may be insufficient due to the influence of combined pathology, concomitant diseases, and the peculiarities of the metabolism of ASMs in patients with epilepsy. Methods: The presented results of experimental and clinical studies of microRNAs (miRs) as epigenetic biomarkers of MetS and AIMetS, which were entered into the different databases, were analyzed for the last decade (2014-2024). Results: A systematic review demonstrated that miRs can act as promising epigenetic biomarkers of key AIMetS domains. However, the results of the review demonstrated the variable role of various miRs and their paralogs in the pathogenesis of AIMetS. Therefore, as part of this study, an miRs signature was proposed that allows us to assess the risk of developing and the severity of AIMetS as low risk, medium risk, and high risk. Conclusions: The mechanisms of development and biomarkers of AIMetS are an actual problem of epileptology, which is still far from being resolved. The development of panels (signatures) of epigenetic biomarkers of this widespread ADR may help to increase the safety of pharmacotherapy of epilepsy. However, to increase the sensitivity and specificity of circulating miRs in the blood as biomarkers of AIMetS, it is necessary to conduct "bridge" studies in order to replicate the results of preclinical and clinical studies into real clinical practice.
Collapse
Affiliation(s)
- Natalia A. Shnayder
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 1 Partizan Zheleznyak St., 660022 Krasnoyarsk, Russia; (D.V.D.); (M.M.P.)
| | - Nikolai A. Pekarets
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
| | - Natalia I. Pekarets
- Department of Psychiatry and Clinical Psychology, Irkutsk State Medical University, 1 Krasny Vosstaniya St., 664003 Irkutsk, Russia;
| | - Diana V. Dmitrenko
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 1 Partizan Zheleznyak St., 660022 Krasnoyarsk, Russia; (D.V.D.); (M.M.P.)
| | - Violetta V. Grechkina
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
| | - Marina M. Petrova
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 1 Partizan Zheleznyak St., 660022 Krasnoyarsk, Russia; (D.V.D.); (M.M.P.)
| | - Mustafa Al-Zamil
- Department of Physiotherapy, Faculty of Continuing Medical Education, Peoples’ Friendship University of Russia, 117198 Moscow, Russia;
| | - Regina F. Nasyrova
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
- Department of Psychiatry, General and Clinical Psychology, Tula State University, 92 Lenin Ave., 300012 Tula, Russia
| |
Collapse
|
2
|
Luo L, Tao FB. Impact of age on cardiometabolic health in children at adiposity rebound: the role of genetic mechanisms. World J Pediatr 2025; 21:252-265. [PMID: 40097891 DOI: 10.1007/s12519-025-00893-8] [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: 11/12/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Identifying effective predictors early in life is crucial to enable timely prevention and intervention to improve cardiometabolic health outcomes. Adiposity rebound (AR) is an important period in early life, with earlier AR increasing the risk of cardiometabolic abnormalities. However, the role and mechanism of genetic factors in this association are unclear. Therefore, this study reviews the potential genetic mechanisms influencing the age at AR, as well as the genetic mechanisms linking earlier AR with cardiometabolic abnormalities. DATA SOURCES A comprehensive literature search was conducted in PubMed and China National Knowledge Infrastructure databases using a combination of medical subject headings terms and related keywords, including "adiposity rebound", "cardiometabolic", "obesity", "BMI trajectory", "diabetes mellitus", "dyslipidemias", "hypertension", "metabolic syndrome", "genetics", and "epigenetic". Citation tracking was performed as a supplementary search strategy. All potentially relevant articles were subsequently subjected to full-text evaluation for eligibility assessment. RESULTS Polymorphisms in the DMRT1, FTO, LEPR, and TFAP2B genes, along with obesity susceptibility, can influence the age at AR. Single-nucleotide polymorphisms associated with the age at AR are enriched in the insulin-like growth factor 1 (IGF-1) signaling pathway, which can be modulated by the LEPR and TFAP2B genes. Shared genetic mechanisms between cardiometabolic abnormalities and the age at AR are influenced by obesity-related genetic variants. These variants regulate the growth hormone (GH)/IGF-1 axis, advancing AR and leading to cardiometabolic abnormalities. Earlier AR alters adiponectin and leptin levels, further activating the GH/IGF-1 axis and creating a vicious cycle. Long-term breastfeeding can counteract the adverse effects of obesity-related genetic susceptibility on AR timing, thereby reducing the genetic risk of cardiometabolic abnormalities. CONCLUSIONS Our results support earlier AR as a marker for identifying cardiometabolic risk and screening high-risk populations at the genetic level.
Collapse
Affiliation(s)
- Ling Luo
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, China
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Anhui Medical University, Hefei, 230032, China
| | - Fang-Biao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, China.
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
3
|
Zhou X, Wu R, Tang G, Shen T, Li W. The predictive function of miR-122-5p and its action mechanism by regulating PKM2 in metabolic syndrome. BMC Endocr Disord 2025; 25:54. [PMID: 40011864 PMCID: PMC11866723 DOI: 10.1186/s12902-025-01888-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/21/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Obesity will cause metabolic syndrome (Mets) easily, and its pathogenesis is not completely clear. AIM To probe into the predictive value of miR-122-5p and its regulatory mechanism in Mets. METHOD The predictive effect of miR-122-5p on Mets was evaluated by constructing a Receiver Operating Characteristic (ROC) curve. The target genes of miR-122-5p were predicted using the ENCORI/starBase and TargetScanHuman databases, and pyruvate kinase M2 (PKM2), closely related to Mets, was screened by GO and KEGG analysis. The roles of miR-122-5p/PKM2 in insulin resistance (IR) were explored by treating the human normal liver cells (HLCs) with palmitic acid (PA) to induce the IR model. The effects of miR-122-5p/PKM2 on glucose metabolism (GM) of HLCs were evaluated by detecting the production of pyruvic acid, lactic acid, and ATP. RESULTS MiR-122-5p was highly expressed in obese people and Mets patients, and its predicted AUC for Mets was 0.876. In HLCs transfected with wild-type PKM2 luciferase vector (PKM2-wt), luciferase activity was attenuated by the miR-122-5p mimic and enhanced by its inhibitor. The expression of PKM2 was inhibited by the miR-122-5p mimic and up-regulated by its inhibitor. The miR-122-5p mimic enhanced PA-induced IR and inhibited the GM of HLCs, which were reversed by overexpression of PKM2. The miR-122-5p inhibitor exerted the opposite effects of its mimic, which were also reversed by silencing of PKM2. CONCLUSION MiR-122-5p, a risk factor for Mets, mediated the IR and abnormal glucose metabolism of HLCs by negatively regulating PKM2. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Xinglu Zhou
- Department of Endocrinology, Shanghai Pudong New Area Gongli Hospital, Shanghai, 200135, China
| | - Rui Wu
- Nursing Department, Geriatric Hospital Affiliated with Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Guangfeng Tang
- Endocrinology Department, The Affiliated Chuzhou Hospital of Anhui Medical University, Chuzhou, 239000, China
| | - Tongtong Shen
- Cardiovascular Medicine, The Affiliated Chuzhou Hospital of Anhui Medical University, No. 369, West Drunken Weng Road, Nanqiao District, Chuzhou, Anhui, 239001, China.
| | - Wei Li
- Department of Endocrinology, Huanggang Central Hospital Affiliated to Yangtze University, No.6, Qi'an Avenue, Huangzhou District, Huanggang, Hubei Province, 438000, China.
| |
Collapse
|
4
|
Sulaiman F, Khyriem C, Dsouza S, Abdul F, Alkhnbashi O, Faraji H, Farooqi M, Al Awadi F, Hassanein M, Ahmed F, Alsharhan M, Tawfik AR, Khamis AH, Bayoumi R. Characterizing Circulating microRNA Signatures of Type 2 Diabetes Subtypes. Int J Mol Sci 2025; 26:637. [PMID: 39859351 PMCID: PMC11766090 DOI: 10.3390/ijms26020637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/04/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Type 2 diabetes (T2D) is a heterogeneous disease influenced by both genetic and environmental factors. Recent studies suggest that T2D subtypes may exhibit distinct gene expression profiles. In this study, we aimed to identify T2D cluster-specific miRNA expression signatures for the previously reported five clinical subtypes that characterize the underlying pathophysiology of long-standing T2D: severe insulin-resistant diabetes (SIRD), severe insulin-deficient diabetes (SIDD), mild age-related diabetes (MARD), mild obesity-related diabetes (MOD), and mild early-onset diabetes (MEOD). We analyzed the circulating microRNAs (miRNAs) in 45 subjects representing the five T2D clusters and 7 non-T2D healthy controls by single-end small RNA sequencing. Bioinformatic analyses identified a total of 430 known circulating miRNAs and 13 previously unreported novel miRNAs. Of these, 71 were upregulated and 37 were downregulated in either controls or individual clusters. Each T2D subtype was associated with a specific dysregulated miRNA profile, distinct from that of healthy controls. Specifically, 3 upregulated miRNAs were unique to SIRD, 1 to MARD, 9 to MOD, and 18 to MEOD. Among the downregulated miRNAs, 11 were specific to SIRD, 9 to SIDD, 2 to MARD, and 1 to MEOD. Our study confirms the heterogeneity of T2D, represented by distinguishable subtypes both clinically and epigenetically and highlights the potential of miRNAs as markers for distinguishing the pathophysiology of T2D subtypes.
Collapse
Affiliation(s)
- Fatima Sulaiman
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| | - Costerwell Khyriem
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| | - Stafny Dsouza
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| | - Fatima Abdul
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| | - Omer Alkhnbashi
- Center for Applied and Translational Genomics, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates;
| | - Hanan Faraji
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| | - Muhammad Farooqi
- Dubai Diabetes Center, Dubai Health, Dubai P.O. Box 7272, United Arab Emirates;
| | - Fatheya Al Awadi
- Endocrinology Department, Dubai Hospital, Dubai Health, Dubai P.O. Box 7272, United Arab Emirates; (F.A.A.); (M.H.)
| | - Mohammed Hassanein
- Endocrinology Department, Dubai Hospital, Dubai Health, Dubai P.O. Box 7272, United Arab Emirates; (F.A.A.); (M.H.)
| | - Fayha Ahmed
- Pathology Department, Dubai Hospital, Dubai Health, Dubai P.O. Box 7272, United Arab Emirates; (F.A.); (M.A.)
| | - Mouza Alsharhan
- Pathology Department, Dubai Hospital, Dubai Health, Dubai P.O. Box 7272, United Arab Emirates; (F.A.); (M.A.)
| | - Abdel Rahman Tawfik
- Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (A.R.T.); (A.H.K.)
| | - Amar Hassan Khamis
- Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (A.R.T.); (A.H.K.)
| | - Riad Bayoumi
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| |
Collapse
|
5
|
Sims SL, Frazier HN, Case SL, Lin RL, Trosper JN, Vekaria HJ, Sullivan PG, Thibault O. Variable bioenergetic sensitivity of neurons and astrocytes to insulin and extracellular glucose. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:33. [PMID: 39524535 PMCID: PMC11549053 DOI: 10.1038/s44324-024-00037-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
Energy flow within cellular elements of the brain is a well-orchestrated, tightly regulated process, however, details underlying these functions at the single-cell level are still poorly understood. Studying hypometabolism in aging and neurodegenerative diseases may benefit from experimentation on unicellular bioenergetics. Here, we examined energy status in neurons and astrocytes using mixed hippocampal cultures and PercevalHR, an ATP:ADP nanosensor. We assessed exposures of several compounds including KCl, glutamate, FCCP, insulin, and glucose. A mitochondrial stress test was performed, and PercevalHR's fluorescence was corrected for pH using pHrodo. Results demonstrate that PercevalHR can reliably report on the energetic status of two cell types that communicate in a mixed-culture setting. While KCl, glutamate, and FCCP showed clear changes in PercevalHR fluorescence, insulin and glucose responses were found to be more subtle and sensitive to extracellular glucose. These results may highlight mechanisms that mediate insulin sensitivity in the brain.
Collapse
Affiliation(s)
- Sophiya L. Sims
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Hilaree N. Frazier
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY USA
| | - Sami L. Case
- Department of Biomedical Sciences, College of Veterinary Medicine & Biomedical Sciences, Colorado State University, Fort Collins, CO USA
| | - Ruei-Lung Lin
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - James N. Trosper
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Hemendra J. Vekaria
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY USA
- Department of Neuroscience, University of Kentucky, Lexington, KY USA
| | - Patrick G. Sullivan
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY USA
- Department of Neuroscience, University of Kentucky, Lexington, KY USA
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY USA
| |
Collapse
|
6
|
Capetini VC, Quintanilha BJ, Garcia BREV, Rogero MM. Dietary modulation of microRNAs in insulin resistance and type 2 diabetes. J Nutr Biochem 2024; 133:109714. [PMID: 39097171 DOI: 10.1016/j.jnutbio.2024.109714] [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: 11/30/2023] [Revised: 07/13/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
The prevalence of type 2 diabetes is increasing worldwide. Various molecular mechanisms have been proposed to interfere with the insulin signaling pathway. Recent advances in proteomics and genomics indicate that one such mechanism involves the post-transcriptional regulation of insulin signaling by microRNA (miRNA). These noncoding RNAs typically induce messenger RNA (mRNA) degradation or translational repression by interacting with the 3' untranslated region (3'UTR) of target mRNA. Dietary components and patterns, which can either enhance or impair the insulin signaling pathway, have been found to regulate miRNA expression in both in vitro and in vivo studies. This review provides an overview of the current knowledge of how dietary components influence the expression of miRNAs related to the control of the insulin signaling pathway and discusses the potential application of these findings in precision nutrition.
Collapse
Affiliation(s)
- Vinícius Cooper Capetini
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil; Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, Institute of Pharmaceutical Science, Department of Pharmacology, King's College London, London, United Kingdom.
| | - Bruna Jardim Quintanilha
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil
| | - Bruna Ruschel Ewald Vega Garcia
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Marcelo Macedo Rogero
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil
| |
Collapse
|
7
|
Umashankar B, Eliasson L, Ooi CY, Kim KW, Shaw JAM, Waters SA. Beyond insulin: Unraveling the complex interplay of ER stress, oxidative damage, and CFTR modulation in CFRD. J Cyst Fibros 2024; 23:842-852. [PMID: 38897882 DOI: 10.1016/j.jcf.2024.06.004] [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: 03/04/2024] [Revised: 05/10/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
CF-related diabetes (CFRD) is a prevalent comorbidity in people with Cystic Fibrosis (CF), significantly impacting morbidity and mortality rates. This review article critically evaluates the current understanding of CFRD molecular mechanisms, including the role of CFTR protein, oxidative stress, unfolded protein response (UPR) and intracellular communication. CFRD manifests from a complex interplay between exocrine pancreatic damage and intrinsic endocrine dysfunction, further complicated by the deleterious effects of misfolded CFTR protein on insulin secretion and action. Studies indicate that ER stress and subsequent UPR activation play critical roles in both exocrine and endocrine pancreatic cell dysfunction, contributing to β-cell loss and insulin insufficiency. Additionally, oxidative stress and altered calcium flux, exacerbated by CFTR dysfunction, impair β-cell survival and function, highlighting the significance of antioxidant pathways in CFRD pathogenesis. Emerging evidence underscores the importance of exosomal microRNAs (miRNAs) in mediating inflammatory and stress responses, offering novel insights into CFRD's molecular landscape. Despite insulin therapy remaining the cornerstone of CFRD management, the variability in response to CFTR modulators underscores the need for personalized treatment approaches. The review advocates for further research into non-CFTR therapeutic targets, emphasizing the need to address the multifaceted pathophysiology of CFRD. Understanding the intricate mechanisms underlying CFRD will pave the way for innovative treatments, moving beyond insulin therapy to target the disease's root causes and improve the quality of life for individuals with CF.
Collapse
Affiliation(s)
- Bala Umashankar
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Lena Eliasson
- Department of Clinical Sciences, Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Scania University Hospital, Malmö, Scania, Sweden
| | - Chee Y Ooi
- Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Department of Gastroenterology, Sydney Children's Hospital Randwick, NSW, Australia
| | - Ki Wook Kim
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Virology and Serology Division (SaViD), New South Wales Health Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
| | - James A M Shaw
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Shafagh A Waters
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
8
|
Cowan E, Sun J, Hamilton A, Ruhrmann S, Karagiannopoulos A, Westholm E, Ofori JK, Luan C, Zhang E, Mulder H, Eliasson L. MicroRNA 29 modulates β-cell mitochondrial metabolism and insulin secretion via underlying miR-29-OXPHOS complex pathways. Acta Physiol (Oxf) 2024; 240:e14180. [PMID: 38801063 DOI: 10.1111/apha.14180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 04/29/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
AIM MicroRNAs (miRNAs) regulate β-cell function, and β-cell mitochondria and insulin secretion are perturbed in diabetes. We aimed to identify key miRNAs regulating β-cell mitochondrial metabolism and novel β-cell miRNA-mitochondrial pathways. METHODS TargetScan (http://www.targetscan.org/) was used to predict if 16 miRNAs implicated in β-cell function target 27 cis-eGenes implicated in mitochondrial activity. The expression of candidate miRNAs and insulin secretion after 24 and 1 h pre-incubation in 2.8, 11.1- and 16.7-mM glucose was measured in clonal INS-1 832/13 β-cells. MiR-29 silenced INS-1 832/13 cells were assessed for insulin secretion (glucose, pyruvate, and K+), target cis-eGene expression (Ndufv3 and Ndufa10 components of mitochondrial complex I (CI)), OXPHOS (CI-V) protein expression, and mitochondrial OXPHOS respiration/activity. The expression of differentially expressed miR-29 miRNAs was evaluated in Goto-Kakizaki (GK) rat, db/db mouse and type 2 diabetic (T2D) human islets, as well as NMRI mouse islets cultured under glucolipotoxic conditions. RESULTS MiR-29, miR-15 and miR-124 were predicted to regulate ~20 cis-eGenes, while miR-29 alone was predicted to regulate ≥12 of these in rat and human species. MiR-29 expression and insulin secretion were reduced in INS-1 832/13 cells after 24 h in elevated glucose. MiR-29 knockdown increased all tested insulin secretory responses, Nudfv3, Ndufa10, complex I and II expression, and cellular mitochondrial OXPHOS. MiR-29 expression was reduced in db/db islets but increased in GK rat and T2D human islets. CONCLUSION We conclude miR-29 is a key miRNA in regulating β-cell mitochondrial metabolism and insulin secretion via underlying miR-29-OXPHOS complex pathways. Furthermore, we infer reduced miR-29 expression compensatorily enhances insulin secretion under glucotoxicity.
Collapse
Affiliation(s)
- E Cowan
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Lund, Sweden
| | - J Sun
- Unit of Molecular Metabolism, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Lund, Sweden
| | - A Hamilton
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Lund, Sweden
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - S Ruhrmann
- Epigenetics and Diabetes Unit, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Lund, Sweden
| | - A Karagiannopoulos
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Lund, Sweden
| | - E Westholm
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Lund, Sweden
| | - J K Ofori
- Epigenetics and Diabetes Unit, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Lund, Sweden
| | - C Luan
- Diabetes-Islet Pathophysiology, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Lund, Sweden
| | - E Zhang
- Diabetes-Islet Pathophysiology, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Lund, Sweden
| | - H Mulder
- Unit of Molecular Metabolism, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Lund, Sweden
| | - L Eliasson
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Lund, Sweden
| |
Collapse
|
9
|
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
|
10
|
Lewis KA, Stroebel BM, Zhang L, Aouizerat B, Mattis AN, Flowers E. MicroRNAs Associated with Metformin Treatment in the Diabetes Prevention Program. Int J Mol Sci 2024; 25:5684. [PMID: 38891870 PMCID: PMC11172132 DOI: 10.3390/ijms25115684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
The Diabetes Prevention Program (DPP) randomized controlled trial demonstrated that metformin treatment reduced progression to type 2 diabetes (T2D) by 31% compared to placebo in adults with prediabetes. Circulating micro-ribonucleic acids (miRs) are promising biomarkers of T2D risk, but little is known about their associations with metformin regimens for T2D risk reduction. We compared the change in 24 circulating miRs from baseline to 2 years in a subset from DPP metformin intervention (n = 50) and placebo (n = 50) groups using Wilcoxon signed rank tests. Spearman correlations were used to evaluate associations between miR change and baseline clinical characteristics. Multiple linear regression was used to adjust for covariates. The sample was 73% female, 17% Black, 13% Hispanic, and 50 ± 11 years. Participants were obese, normotensive, prediabetic, and dyslipidemic. Change in 12 miR levels from baseline to 2 years was significantly different in the metformin group compared with placebo after adjusting for multiple comparisons: six (let-7c-5p, miR-151a-3p, miR-17-5p, miR-20b-5p, miR-29b-3p, and miR-93-5p) were significantly upregulated and six (miR-130b-3p, miR-22-3p, miR-222-3p, miR-320a-3p, miR-320c, miR-92a-3p) were significantly downregulated in the metformin group. These miRs help to explain how metformin is linked to T2D risk reduction, which may lead to novel biomarkers, therapeutics, and precision health strategies.
Collapse
Affiliation(s)
- Kimberly A. Lewis
- Department of Physiological Nursing, School of Nursing, University of California, 2 Koret Way, San Francisco, CA 94143, USA; (B.M.S.); (E.F.)
| | - Benjamin M. Stroebel
- Department of Physiological Nursing, School of Nursing, University of California, 2 Koret Way, San Francisco, CA 94143, USA; (B.M.S.); (E.F.)
| | - Li Zhang
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94143, USA;
| | | | - Aras N. Mattis
- Department of Pathology, University of California, San Francisco, CA 94143, USA;
| | - Elena Flowers
- Department of Physiological Nursing, School of Nursing, University of California, 2 Koret Way, San Francisco, CA 94143, USA; (B.M.S.); (E.F.)
- Institute for Human Genetics, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
11
|
Li X, Sun M, Wang Z, Sun S, Wang Y. Recent advances in mechanistic studies of heart failure with preserved ejection fraction and its comorbidities-Role of microRNAs. Eur J Clin Invest 2024; 54:e14130. [PMID: 38071416 DOI: 10.1111/eci.14130] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/22/2023] [Accepted: 11/11/2023] [Indexed: 02/15/2024]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a multifaceted syndrome with a complex aetiology commonly associated with comorbidities such as diabetes mellitus, obesity, hypertension and renal disease. Various diseases induce systemic, chronic and low-grade inflammation; microvascular dysfunction; metabolic stress; tissue ischemia; and fibrosis, leading to HFpEF. An effective treatment for HFpEF is lacking, largely owing to its pathophysiological heterogeneity. Recent studies have revealed that microRNAs (miRNAs) play crucial roles in regulating the pathogenesis of HFpEF and its comorbidities. METHODS This narrative review included original articles and reviews published over the past 20 years found through 'PubMed' and 'Web of Science'. The search terms included "HFpEF," "MicroRNAs," "comorbidities," "Microvascular Dysfunction (MVD)," "inflammation," "pathophysiology," "endothelial dysfunction," "energy metabolism abnormalities" "cardiac fibrosis" and "treatment." RESULTS Inflammation, MVD, abnormal energy metabolism, myocardial hypertrophy and myocardial fibrosis are important pathophysiological mechanisms underlying HFpEF. As gene expression regulators, miRNAs may contribute to the pathophysiology of HFpEF and are expected to serve in the stratification of patients with HFpEF and as prognostic indicators for monitoring treatment responses. CONCLUSIONS A customized strategy based on miRNAs has emerged as an effective treatment for HFpEF. In this review, we discuss recent research surrounding miRNAs and HFpEF and propose potential miRNA targets for the pathophysiology of HFpEF and its comorbidities. Although current research concerning miRNAs and their therapeutic potential is in its early stages, miRNA-based diagnostics and therapeutics hold great promise in the future.
Collapse
Affiliation(s)
- Xiaonan Li
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| | - Min Sun
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| | - Zhe Wang
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| | - Siming Sun
- Department of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Yuehui Wang
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Lewis KA, Stroebel B, Zhang L, Aouizerat B, Mattis A, Flowers E. MicroRNAs Associated with Metformin Treatment in the Diabetes Prevention Program. RESEARCH SQUARE 2024:rs.3.rs-3846347. [PMID: 38313262 PMCID: PMC10836103 DOI: 10.21203/rs.3.rs-3846347/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
The Diabetes Prevention Program (DPP) randomized controlled trial demonstrated that metformin treatment reduced progression to type 2 diabetes (T2D) by 31% compared to placebo in adults with prediabetes. Circulating micro-ribonucleic acids (miRs) are promising biomarkers of T2D risk, but little is known about their associations with metformin regimens for T2D risk reduction. We compared the change in 24 circulating miRs from baseline to 2 years in a subset from DPP metformin intervention (n = 50) and placebo (n = 50) groups using Wilcoxon signed rank tests. Spearman's correlations were used to evaluate associations between miR change and baseline clinical characteristics. Multiple linear regression was used to adjust for covariates. The sample was 73% female, 17% Black, 13% Hispanic, and 50 ± 11 years. Participants were obese, normotensive, prediabetic, and dyslipidemic. Change in 12 miR levels from baseline to 2 years was significantly different in the metformin group compared with placebo after adjusting for multiple comparisons: six (let-7c-5p, miR-151a-3p, miR-17-5p, miR-20b-5p, miR-29b-3p, and miR-93-5p) were significantly upregulated and six (miR-130b-3p, miR-22-3p, miR-222-3p, miR-320a-3p, miR-320c, miR-92a-3p) were significantly downregulated in the metformin group. These miRs help to explain how metformin is linked to T2D risk reduction, which may lead to novel biomarkers, therapeutics, and precision-health strategies.
Collapse
Affiliation(s)
| | | | - Li Zhang
- University of California San Francisco
| | | | | | | |
Collapse
|
13
|
Kovac L, Speckmann T, Jähnert M, Gottmann P, Fritsche L, Häring HU, Birkenfeld AL, Fritsche A, Schürmann A, Ouni M. Identification of MicroRNAs Associated with Prediabetic Status in Obese Women. Int J Mol Sci 2023; 24:15673. [PMID: 37958657 PMCID: PMC10648886 DOI: 10.3390/ijms242115673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
MicroRNAs (miRNAs) recently emerged as means of communication between insulin-sensitive tissues to mediate diabetes development and progression, and as such they present a valuable proxy for epigenetic alterations associated with type 2 diabetes. In order to identify miRNA markers for the precursor of diabetes called prediabetes, we applied a translational approach encompassing analysis of human plasma samples, mouse tissues and an in vitro validation system. MiR-652-3p, miR-877-5p, miR-93-5p, miR-130a-3p, miR-152-3p and let-7i-5p were increased in plasma of women with impaired fasting glucose levels (IFG) compared to those with normal fasting glucose and normal glucose tolerance (NGT). Among these, let-7i-5p and miR-93-5p correlated with fasting blood glucose levels. Human data were then compared to miRNome data obtained from islets of Langerhans and adipose tissue of 10-week-old female New Zealand Obese mice, which differ in their degree of hyperglycemia and liver fat content. Similar to human plasma, let-7i-5p was increased in adipose tissue and islets of Langerhans of diabetes-prone mice. As predicted by the in silico analysis, overexpression of let-7i-5p in the rat β-cell line INS-1 832/12 resulted in downregulation of insulin signaling pathway components (Insr, Rictor, Prkcb, Clock, Sos1 and Kcnma1). Taken together, our integrated approach highlighted let-7i-5p as a potential regulator of whole-body insulin sensitivity and a novel marker of prediabetes in women.
Collapse
Affiliation(s)
- Leona Kovac
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (L.K.); (M.J.); (P.G.); (M.O.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (L.F.); (H.-U.H.); (A.L.B.); (A.F.)
| | - Thilo Speckmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (L.K.); (M.J.); (P.G.); (M.O.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (L.F.); (H.-U.H.); (A.L.B.); (A.F.)
| | - Markus Jähnert
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (L.K.); (M.J.); (P.G.); (M.O.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (L.F.); (H.-U.H.); (A.L.B.); (A.F.)
| | - Pascal Gottmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (L.K.); (M.J.); (P.G.); (M.O.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (L.F.); (H.-U.H.); (A.L.B.); (A.F.)
| | - Louise Fritsche
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (L.F.); (H.-U.H.); (A.L.B.); (A.F.)
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, University of Tübingen, 72074 Tübingen, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (L.F.); (H.-U.H.); (A.L.B.); (A.F.)
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, University of Tübingen, 72074 Tübingen, Germany
- Department of Internal Medicine IV, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Andreas L. Birkenfeld
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (L.F.); (H.-U.H.); (A.L.B.); (A.F.)
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, University of Tübingen, 72074 Tübingen, Germany
- Department of Internal Medicine IV, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Andreas Fritsche
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (L.F.); (H.-U.H.); (A.L.B.); (A.F.)
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, University of Tübingen, 72074 Tübingen, Germany
- Department of Internal Medicine IV, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (L.K.); (M.J.); (P.G.); (M.O.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (L.F.); (H.-U.H.); (A.L.B.); (A.F.)
- Institute of Nutritional Sciences, University of Potsdam, 14558 Nuthetal, Germany
| | - Meriem Ouni
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (L.K.); (M.J.); (P.G.); (M.O.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (L.F.); (H.-U.H.); (A.L.B.); (A.F.)
| |
Collapse
|
14
|
Morishita A, Oura K, Tadokoro T, Fujita K, Tani J, Kobara H, Ono M, Himoto T, Masaki T. MicroRNAs and Nonalcoholic Steatohepatitis: A Review. Int J Mol Sci 2023; 24:14482. [PMID: 37833930 PMCID: PMC10572537 DOI: 10.3390/ijms241914482] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a clinicopathologic syndrome caused by fat deposition in hepatocytes. Patients with nonalcoholic steatohepatitis (NASH), an advanced form of NAFLD with severe fibrosis, are at high risk for liver-related complications, including hepatocellular carcinoma (HCC). However, the mechanism of progression from simple fat deposition to NASH is complex, and previous reports have linked NAFLD to gut microbiota, bile acids, immunity, adipokines, oxidative stress, and genetic or epigenetic factors. NASH-related liver injury involves multiple cell types, and intercellular signaling is thought to be mediated by extracellular vesicles. MicroRNAs (miRNAs) are short, noncoding RNAs that play important roles as post-transcriptional regulators of gene expression and have been implicated in the pathogenesis of various diseases. Recently, many reports have implicated microRNAs in the pathogenesis of NALFD/NASH, suggesting that exosomal miRNAs are potential non-invasive and sensitive biomarkers and that the microRNAs involved in the mechanism of the progression of NASH may be potential therapeutic target molecules. We are interested in which miRNAs are involved in the pathogenesis of NASH and which are potential target molecules for therapy. We summarize targeted miRNAs associated with the etiology and progression of NASH and discuss each miRNA in terms of its pathophysiology, potential therapeutic applications, and efficacy as a NASH biomarker.
Collapse
Affiliation(s)
| | | | - Tomoko Tadokoro
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kita-gun 761-0793, Japan; (A.M.); (K.O.); (K.F.); (J.T.); (H.K.); (M.O.); (T.H.); (T.M.)
| | | | | | | | | | | | | |
Collapse
|
15
|
Fu D, Zhao H, Huang Y, Li J, Feng H, Li A, Liu Y, He L. Metformin regulates the effects of IR and IGF-1R methylation on mast cell activation and airway reactivity in diabetic rats with asthma through miR-152-3p/DNMT1 axis. Cell Biol Toxicol 2023; 39:1851-1872. [PMID: 36547818 DOI: 10.1007/s10565-022-09787-1] [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: 07/28/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND/AIM Metformin is a drug for treating type 2 diabetes mellitus (T2DM). Recently, metformin has been shown to reduce the risks of asthma-associated outcomes and asthma deterioration, thereby holding promise as a superior medicine for diabetic patients with asthma. However, the mechanism by which metformin reduces diabetic asthma is yet to be clarified. This study aimed at ascertaining the downstream molecules underlying the effect of metformin on the activation of mast cells (MCs) and airway reactivity in a concomitant diabetic and asthmatic rat model. METHODS A T2DM model was induced utilizing a high-fat diet and streptozotocin. Then, 10% ovalbumin was utilized to stimulate asthma-like pathology in the T2DM rats. RBL-2H3 cells were induced by anti-dinitrophenyl-specific immunoglobulin E for constructing an in vitro model. Luciferase assay and RNA immunoprecipitation (IP) assay were conducted to identify the interaction between microRNA-152-3p (miR-152-3p) and DNA methyltransferase 1 (DNMT1), while chromatin IP to identify the binding of DNMT1 to insulin receptor (IR) and insulin-like growth factor 1 receptor (IGF-1R) promoters. The effects of metformin on both pathological changes in vivo and biological behaviors of cells were evaluated. Using gain- and loss-of-function approaches, we assessed the role of the two interactions in the metformin-induced effect. RESULTS It was suggested that metformin could impede the MC activation and airway resistance in the concomitant diabetic and asthmatic rats. Additionally, metformin downregulated IR and IGF-1R through DNMT1-dependent methylation to repress MC activation and airway resistance. DNMT1 was testified to be a target gene of miR-152-3p. Furthermore, miR-152-3p-induced silencing of DNMT1 was blocked by metformin, hence restraining MC activation and airway resistance. CONCLUSION The findings cumulatively demonstrate that metformin downregulates IR/IGF-1R to block MC activation and airway resistance via impairing the binding affinity between miR-152-3p and DNMT1.
Collapse
Affiliation(s)
- Dan Fu
- Department of Endocrinology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Hailu Zhao
- Diabetic Systems Center, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi, 541000, People's Republic of China
| | - Yan Huang
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China
| | - Jingjuan Li
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China
| | - Huafeng Feng
- Department of Anesthesiology, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People's Republic of China
| | - Aiguo Li
- Department of Anesthesiology, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People's Republic of China
| | - Yefen Liu
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China
| | - Liang He
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China.
| |
Collapse
|
16
|
Karuga FF, Jaromirska J, Malicki M, Sochal M, Szmyd B, Białasiewicz P, Strzelecki D, Gabryelska A. The role of microRNAs in pathophysiology and diagnostics of metabolic complications in obstructive sleep apnea patients. Front Mol Neurosci 2023; 16:1208886. [PMID: 37547923 PMCID: PMC10403239 DOI: 10.3389/fnmol.2023.1208886] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/04/2023] [Indexed: 08/08/2023] Open
Abstract
Obstructive sleep apnea (OSA) is one of the most common sleep disorders, which is characterized by recurrent apneas and/or hypopneas occurring during sleep due to upper airway obstruction. Among a variety of health consequences, OSA patients are particularly susceptible to developing metabolic complications, such as metabolic syndrome and diabetes mellitus type 2. MicroRNAs (miRNAs) as epigenetic modulators are promising particles in both understanding the pathophysiology of OSA and the prediction of OSA complications. This review describes the role of miRNAs in the development of OSA-associated metabolic complications. Moreover, it summarizes the usefulness of miRNAs as biomarkers in predicting the aforementioned OSA complications.
Collapse
Affiliation(s)
- Filip Franciszek Karuga
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| | - Julia Jaromirska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| | - Mikołaj Malicki
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| | - Bartosz Szmyd
- Department of Neurosurgery and Neuro-Oncology, Barlicki University Hospital, Medical University of Lodz, Lodz, Poland
- Department of Pediatrics, Oncology, and Hematology, Medical University of Lodz, Lodz, Poland
| | - Piotr Białasiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Lodz, Poland
| | - Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
17
|
Chaudhari U, Pohjolainen L, Ruskoaho H, Talman V. Genome-wide profiling of miRNA-gene regulatory networks in mouse postnatal heart development-implications for cardiac regeneration. Front Cardiovasc Med 2023; 10:1148618. [PMID: 37283582 PMCID: PMC10241105 DOI: 10.3389/fcvm.2023.1148618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/02/2023] [Indexed: 06/08/2023] Open
Abstract
Background After birth, mammalian cardiomyocytes substantially lose proliferative capacity with a concomitant switch from glycolytic to oxidative mitochondrial energy metabolism. Micro-RNAs (miRNAs) regulate gene expression and thus control various cellular processes. Their roles in the postnatal loss of cardiac regeneration are however still largely unclear. Here, we aimed to identify miRNA-gene regulatory networks in the neonatal heart to uncover role of miRNAs in regulation of cell cycle and metabolism. Methods and results We performed global miRNA expression profiling using total RNA extracted from mouse ventricular tissue samples collected on postnatal day 1 (P01), P04, P09, and P23. We used the miRWalk database to predict the potential target genes of differentially expressed miRNAs and our previously published mRNA transcriptomics data to identify verified target genes that showed a concomitant differential expression in the neonatal heart. We then analyzed the biological functions of the identified miRNA-gene regulatory networks using enriched Gene Ontology (GO) and KEGG pathway analyses. Altogether 46 miRNAs were differentially expressed in the distinct stages of neonatal heart development. For twenty miRNAs, up- or downregulation took place within the first 9 postnatal days thus correlating temporally with the loss of cardiac regeneration. Importantly, for several miRNAs, including miR-150-5p, miR-484, and miR-210-3p there are no previous reports about their role in cardiac development or disease. The miRNA-gene regulatory networks of upregulated miRNAs negatively regulated biological processes and KEGG pathways related to cell proliferation, while downregulated miRNAs positively regulated biological processes and KEGG pathways associated with activation of mitochondrial metabolism and developmental hypertrophic growth. Conclusion This study reports miRNAs and miRNA-gene regulatory networks with no previously described role in cardiac development or disease. These findings may help in elucidating regulatory mechanism of cardiac regeneration and in the development of regenerative therapies.
Collapse
|
18
|
Westholm E, Edlund A, Karagiannopoulos A, Wendt A, Eliasson L. Interleukin-4 reduces insulin secretion in human islets from healthy but not type-2 diabetic donors. Biochem Biophys Res Commun 2023; 649:87-92. [PMID: 36758483 DOI: 10.1016/j.bbrc.2023.01.092] [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: 01/13/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023]
Abstract
Type 2 diabetes (T2D) is associated with low-grade inflammation. Here we investigate if the anti-inflammatory cytokine interleukin-4 (IL-4) affects glucose-stimulated insulin secretion (GSIS) in human islets from non-diabetic (ND) and type-2 diabetic (T2D) donors. We first confirmed that GSIS is reduced in islets from T2D donors. Treatment with IL-4 for 48 h had no further effect on GSIS in these islets but significantly reduced secretion in ND islets. Acute treatment with IL-4 for 1 h had no effect on GSIS in ND islets which led us to suspect that IL-4 affects a slow cellular mechanism such as gene transcription. IL-4 has been reported to regulate miR-378a-3p and, indeed, we found that this microRNA was increased with IL-4 treatment. However, overexpression of miR-378a-3p in the human beta cell line EndoC-βH1 did not affect GSIS. MiR-378a-3p is transcribed from the same gene as peroxisome proliferator-activated receptor gamma co-activator 1 beta (PCG-1β) and we found that IL-4 treatment showed a clear tendency to increased gene expression of PCG-1β. PCG-1β is a co-activator of peroxisome proliferator-activated receptor gamma (PPARγ) and, the gene expression of PPARγ was also increased with IL-4 treatment. Our data suggests that the protective role of IL-4 on beta cell survival comes at the cost of lowered insulin secretion, presumably involving the PPARγ-pathway.
Collapse
Affiliation(s)
- Efraim Westholm
- Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences in Malmö, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Anna Edlund
- Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences in Malmö, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Alexandros Karagiannopoulos
- Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences in Malmö, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Anna Wendt
- Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences in Malmö, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Lena Eliasson
- Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences in Malmö, Lund University, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
19
|
Nagao M, Asai A, Eliasson L, Oikawa S. Selectively bred rodent models for studying the etiology of type 2 diabetes: Goto-Kakizaki rats and Oikawa-Nagao mice. Endocr J 2023; 70:19-30. [PMID: 36477370 DOI: 10.1507/endocrj.ej22-0253] [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: 12/12/2022] Open
Abstract
Type 2 diabetes (T2D) is a polygenic disease and studies to understand the etiology of the disease have required selectively bred animal models with polygenic background. In this review, we present two models; the Goto-Kakizaki (GK) rat and the Oikawa-Nagao Diabetes-Prone (ON-DP) and Diabetes-Resistant (ON-DR) mouse. The GK rat was developed by continuous selective breeding for glucose tolerance from the outbred Wistar rat around 50 years ago. The main cause of spontaneous hyperglycemia in this model is insulin secretion deficiency from pancreatic β-cells and mild insulin resistance in insulin target organs. A disadvantage of the GK rat is that environmental factors have not been considered in the selective breeding. Hence, the GK rat may not be suitable for elucidating predisposition to diabetes under certain environmental conditions, such as a high-fat diet. Therefore, we recently established two mouse lines with different susceptibilities to diet-induced diabetes, which are prone and resistant to the development of diabetes, designated as the ON-DP and ON-DR mouse, respectively. The two ON mouse lines were established by continuous selective breeding for inferior and superior glucose tolerance after high-fat diet feeding in hybrid mice of three inbred strains. Studies of phenotypic differences between ON-DP and ON-DR mice and their underlying molecular mechanisms will shed light on predisposing factors for the development of T2D in the modern obesogenic environment. This review summarizes the background and the phenotypic differences and similarities of GK rats and ON mice and highlights the advantages of using selectively bred rodent models in diabetes research.
Collapse
Affiliation(s)
- Mototsugu Nagao
- Department of Endocrinology, Metabolism and Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö 214 28, Sweden
- Clincal Research Centre (CRC), Skåne University Hospital(SUS), Malmö 214 28, Sweden
| | - Akira Asai
- Department of Endocrinology, Metabolism and Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Lena Eliasson
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö 214 28, Sweden
- Clincal Research Centre (CRC), Skåne University Hospital(SUS), Malmö 214 28, Sweden
| | - Shinichi Oikawa
- Department of Endocrinology, Metabolism and Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| |
Collapse
|
20
|
Nigam S, Moore A, Wang P. miRNA Theranostic Nanoparticles Promote Pancreatic Beta Cell Proliferation in Type 1 Diabetes Model. Methods Mol Biol 2023; 2592:207-218. [PMID: 36507996 DOI: 10.1007/978-1-0716-2807-2_15] [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] [Indexed: 12/14/2022]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disorder which affects the insulin-producing beta cells in the pancreas. A variety of strategies, namely, insulin replacement therapy, engineered vaccines, immunomodulators, etc., have been explored to correct this condition. Recent studies have attributed the development of T1D to the anomalous expression of microRNAs in the pancreatic islets. Here, we describe the protocol for the development of a theranostic approach to modify the expression of aberrant miRNAs. The MRI-based nanodrug consists of superparamagnetic iron oxide nanoparticles conjugated to microRNA-targeting oligonucleotides that can promote proliferation of pancreatic beta cells in a mouse model of T1D. This theranostic approach can successfully serve as a potential therapeutic approach for the targeted treatment of T1D with minimal side effects.
Collapse
Affiliation(s)
- Saumya Nigam
- Precision Health Program, Michigan State University, East Lansing, MI, USA
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Anna Moore
- Precision Health Program, Michigan State University, East Lansing, MI, USA.
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA.
| | - Ping Wang
- Precision Health Program, Michigan State University, East Lansing, MI, USA.
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
21
|
Circulating microRNA levels differ in the early stages of insulin resistance in prepubertal children with obesity. Life Sci 2022; 312:121246. [PMID: 36455651 PMCID: PMC10375861 DOI: 10.1016/j.lfs.2022.121246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
AIMS The increasing prevalence of childhood obesity escalates the risk for related complications. Circulating microRNAs (miRNAs) have been suggested as good predictive markers of insulin resistance in those with obesity. The aim was to identify a circulating miRNA profile that reflects insulin resistance in prepubertal children with obesity. MATERIAL AND METHODS Plasma miRNAs were measured in prepubertal children (n = 63, 5-9 years) using TaqMan Advanced miRNA Human Serum/Plasma plates and then were validated by RT-qPCR. Subjects were divided into normal weight (n = 20, NW) and overweight or obese (n = 43, OW/OB) groups according to their BMI z-scores. The OW/OB group was further subdivided into insulin sensitive or metabolically healthy obese (n = 26, MHO) and insulin resistant or metabolically unhealthy obese (n = 17, MUO) according to HOMA-IR. KEY FINDINGS While no differences were observed in the fasting plasma glucose levels, serum insulin levels were significantly elevated in the OW/OB compared to the NW group. Of 188 screened miRNAs, eleven were differentially expressed between the NW and OW/OB groups. Validation confirmed increased circulating levels of miR-146a-5p and miR-18a-5p in the OW/OB group, which correlated with BMI z-score. Interestingly, miR-146a-5p was also correlated with HOMA-IR index. While only miR-18a-5p was upregulated in the OW/OB children, independently of their degree of insulin sensitivity, miR-146-5p, miR-423-3p and miR-152-3p were associated with insulin resistance. SIGNIFICANCE The present study provides evidence of molecular alterations that occur early in life in prepubertal obesity. These alterations may potentially be crucial for targeted prevention or prompt precision therapeutic development and subsequent interventions.
Collapse
|
22
|
Analysis of serum circulating MicroRNAs level in Malaysian patients with gestational diabetes mellitus. Sci Rep 2022; 12:20295. [PMID: 36434110 PMCID: PMC9700700 DOI: 10.1038/s41598-022-23816-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 11/07/2022] [Indexed: 11/26/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is a severe global issue that requires immediate attention. MicroRNA expression abnormalities are possibly disease-specific and may contribute to GDM pathological processes. To date, there is limited data on miRNA profiling in GDM, especially that involves a longitudinal study. Here, we performed miRNA expression profiling in the entire duration of pregnancy (during pregnancy until parturition and postpartum) using a miRNA- polymerase chain reaction array (miRNA-PCRArray) and in-silico analysis to identify unique miRNAs expression and their anticipated target genes in Malay maternal serum. MiRNA expression levels and their unique potential as biomarkers were explored in this work. In GDM patients, the expression levels of hsa-miR-193a, hsa-miR-21, hsa-miR-23a, and hsa-miR-361 were significantly increased, but miR-130a was significantly downregulated. The area under the curve (AUC) and receiver operating characteristic (ROC) curve study demonstrated that hsa-miR-193a (AUC = 0.89060 ± 04,470, P = 0.0001), hsa-miR-21 (AUC = 0.89500 ± 04,411, P = 0.0001), and miR-130a (AUC = 0.6939 ± 0.05845, P = 0.0025) had potential biomarker features in GDM. In-silico analysis also revealed that KLF (Kruppel-Like family of transcription factor), ZNF25 (Zinc finger protein 25), AFF4 (ALF transcription elongation factor 4), C1orf143 (long intergenic non-protein coding RNA 2869), SRSF2 (serine and arginine rich splicing factor 2), and ZNF655 (Zinc finger protein 655) were prominent genes targeted by the common nodes of miR23a, miR130, miR193a, miR21, and miR361.Our findings suggest that circulating microRNAs in the first trimester has the potential for GDM screening in the Malay population.
Collapse
|
23
|
Palihaderu PADS, Mendis BILM, Premarathne JMKJK, Dias WKRR, Yeap SK, Ho WY, Dissanayake AS, Rajapakse IH, Karunanayake P, Senarath U, Satharasinghe DA. Therapeutic Potential of miRNAs for Type 2 Diabetes Mellitus: An Overview. Epigenet Insights 2022; 15:25168657221130041. [PMID: 36262691 PMCID: PMC9575458 DOI: 10.1177/25168657221130041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/14/2022] [Indexed: 11/05/2022] Open
Abstract
MicroRNA(miRNA)s have been identified as an emerging class for therapeutic
interventions mainly due to their extracellularly stable presence in humans and
animals and their potential for horizontal transmission and action. However,
treating Type 2 diabetes mellitus using this technology has yet been in a
nascent state. MiRNAs play a significant role in the pathogenesis of Type 2
diabetes mellitus establishing the potential for utilizing miRNA-based
therapeutic interventions to treat the disease. Recently, the administration of
miRNA mimics or antimiRs in-vivo has resulted in positive modulation of glucose
and lipid metabolism. Further, several cell culture-based interventions have
suggested beta cell regeneration potential in miRNAs. Nevertheless, few such
miRNA-based therapeutic approaches have reached the clinical phase. Therefore,
future research contributions would identify the possibility of miRNA
therapeutics for tackling T2DM. This article briefly reported recent
developments on miRNA-based therapeutics for treating Type 2 Diabetes mellitus,
associated implications, gaps, and recommendations for future studies.
Collapse
Affiliation(s)
- PADS Palihaderu
- Department of Basic Veterinary
Sciences, Faculty of Veterinary Medicine and Animal Science, University of
Peradeniya, Peradeniya, Sri Lanka
| | - BILM Mendis
- Department of Basic Veterinary
Sciences, Faculty of Veterinary Medicine and Animal Science, University of
Peradeniya, Peradeniya, Sri Lanka
| | - JMKJK Premarathne
- Department of Livestock and Avian
Sciences, Faculty of Livestock, Fisheries, and Nutrition, Wayamba University of Sri
Lanka, Makandura, Gonawila (NWP), Sri Lanka
| | - WKRR Dias
- Department of North Indian Music,
Faculty of Music, University of the Visual and Performing Arts, Colombo, Sri
Lanka
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences,
Xiamen University Malaysia Campus, Jalan Sunsuria, Bandar Sunsuria, Sepang,
Selangor, Malaysia
| | - Wan Yong Ho
- Division of Biomedical Sciences,
Faculty of Medicine and Health Sciences, University of Nottingham (Malaysia Campus),
Semenyih, Malaysia
| | - AS Dissanayake
- Department of Clinical Medicine,
Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | - IH Rajapakse
- Department of Psychiatry, Faculty of
Medicine, University of Ruhuna, Galle, Sri Lanka
| | - P Karunanayake
- Department of Clinical Medicine,
Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - U Senarath
- Department of Community Medicine,
Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - DA Satharasinghe
- Department of Basic Veterinary
Sciences, Faculty of Veterinary Medicine and Animal Science, University of
Peradeniya, Peradeniya, Sri Lanka,DA Satharasinghe, Department of Basic
Veterinary Sciences, Faculty of Veterinary Medicine and Animal Science,
University of Peradeniya, Peradeniya, 20400, Sri Lanka.
| |
Collapse
|
24
|
Pandur E, Szabó I, Hormay E, Pap R, Almási A, Sipos K, Farkas V, Karádi Z. Alterations of the expression levels of glucose, inflammation, and iron metabolism related miRNAs and their target genes in the hypothalamus of STZ-induced rat diabetes model. Diabetol Metab Syndr 2022; 14:147. [PMID: 36210435 PMCID: PMC9549668 DOI: 10.1186/s13098-022-00919-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The hypothalamus of the central nervous system is implicated in the development of diabetes due to its glucose-sensing function. Dysregulation of the hypothalamic glucose-sensing neurons leads to abnormal glucose metabolism. It has been described that fractalkine (FKN) is involved in the development of hypothalamic inflammation, which may be one of the underlying causes of a diabetic condition. Moreover, iron may play a role in the pathogenesis of diabetes via the regulation of hepcidin, the iron regulatory hormone synthesis. MicroRNAs (miRNAs) are short non-coding molecules working as key regulators of gene expression, usually by inhibiting translation. Hypothalamic miRNAs are supposed to have a role in the control of energy balance by acting as regulators of hypothalamic glucose metabolism via influencing translation. METHODS Using a miRNA array, we analysed the expression of diabetes, inflammation, and iron metabolism related miRNAs in the hypothalamus of a streptozotocin-induced rat type 1 diabetes model. Determination of the effect of miRNAs altered by STZ treatment on the target genes was carried out at protein level. RESULTS We found 18 miRNAs with altered expression levels in the hypothalamus of the STZ-treated animals, which act as the regulators of mRNAs involved in glucose metabolism, pro-inflammatory cytokine synthesis, and iron homeostasis suggesting a link between these processes in diabetes. The alterations in the expression level of these miRNAs could modify hypothalamic glucose sensing, tolerance, uptake, and phosphorylation by affecting the stability of hexokinase-2, insulin receptor, leptin receptor, glucokinase, GLUT4, insulin-like growth factor receptor 1, and phosphoenolpyruvate carboxykinase mRNA molecules. Additional miRNAs were found to be altered resulting in the elevation of FKN protein. The miRNA, mRNA, and protein analyses of the diabetic hypothalamus revealed that the iron import, export, and iron storage were all influenced by miRNAs suggesting the disturbance of hypothalamic iron homeostasis. CONCLUSION It can be supposed that glucose metabolism, inflammation, and iron homeostasis of the hypothalamus are linked via the altered expression of common miRNAs as well as the increased expression of FKN, which contribute to the imbalance of energy homeostasis, the synthesis of pro-inflammatory cytokines, and the iron accumulation of the hypothalamus. The results raise the possibility that FKN could be a potential target of new therapies targeting both inflammation and iron disturbances in diabetic conditions.
Collapse
Affiliation(s)
- Edina Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 4., 7624, Pécs, Hungary.
| | - István Szabó
- Institute of Physiology, Medical School, University of Pécs, Szigeti út 12., 7624, Pécs, Hungary
| | - Edina Hormay
- Institute of Physiology, Medical School, University of Pécs, Szigeti út 12., 7624, Pécs, Hungary
| | - Ramóna Pap
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 4., 7624, Pécs, Hungary
| | - Attila Almási
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Pécs, Rókus u. 4., 7624, Pécs, Hungary
| | - Katalin Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 4., 7624, Pécs, Hungary
| | - Viktória Farkas
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 4., 7624, Pécs, Hungary
| | - Zoltán Karádi
- Institute of Physiology, Medical School, University of Pécs, Szigeti út 12., 7624, Pécs, Hungary
| |
Collapse
|
25
|
Chen MY, Meng XF, Han YP, Yan JL, Xiao C, Qian LB. Profile of crosstalk between glucose and lipid metabolic disturbance and diabetic cardiomyopathy: Inflammation and oxidative stress. Front Endocrinol (Lausanne) 2022; 13:983713. [PMID: 36187088 PMCID: PMC9521548 DOI: 10.3389/fendo.2022.983713] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, the risk, such as hypertension, obesity and diabetes mellitus, of cardiovascular diseases has been increasing explosively with the development of living conditions and the expansion of social psychological pressure. The disturbance of glucose and lipid metabolism contributes to both collapse of myocardial structure and cardiac dysfunction, which ultimately leads to diabetic cardiomyopathy. The pathogenesis of diabetic cardiomyopathy is multifactorial, including inflammatory cascade activation, oxidative/nitrative stress, and the following impaired Ca2+ handling induced by insulin resistance/hyperinsulinemia, hyperglycemia, hyperlipidemia in diabetes. Some key alterations of cellular signaling network, such as translocation of CD36 to sarcolemma, activation of NLRP3 inflammasome, up-regulation of AGE/RAGE system, and disequilibrium of micro-RNA, mediate diabetic oxidative stress/inflammation related myocardial remodeling and ventricular dysfunction in the context of glucose and lipid metabolic disturbance. Here, we summarized the detailed oxidative stress/inflammation network by which the abnormality of glucose and lipid metabolism facilitates diabetic cardiomyopathy.
Collapse
Affiliation(s)
| | | | | | | | - Chi Xiao
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Ling-Bo Qian
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
26
|
Mendonca A, Thandapani P, Nagarajan P, Venkatesh S, Sundaresan S. Role of microRNAs in regulation of insulin secretion and insulin signaling involved in type 2 diabetes mellitus. J Biosci 2022. [DOI: 10.1007/s12038-022-00295-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
27
|
Heris HV, Zahraei Z. miRNAs: Regulators of immune system in diabetes. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2022; 790:108442. [PMID: 36089265 DOI: 10.1016/j.mrrev.2022.108442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 01/01/2023]
Abstract
Diabetes, one of the most common multifactorial metabolic disorders, is a jeopardizing cause of human health worldwide. MicroRNAs (miRNAs) are a group of small non-coding RNAs that have been contributed to the regulation of gene expression through post-transcriptional mechanisms. The potential role of miRNAs has been studied in the most of biological processes and mechanisms underlying the progression of variety diseases including diabetes. In this review, we focus on the role of miRNAs in regulating pivotal molecular and cellular mechanisms associated with immune system that progress diabetic disorders.
Collapse
Affiliation(s)
- Helaleh Vaezi Heris
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Zohreh Zahraei
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Iran.
| |
Collapse
|
28
|
Li W, Feng Q, Wang C, Yin Z, Li X, Li L. LncXIST Facilitates Iron Overload and Iron Overload-Induced Islet Beta Cell Injury in Type 2 Diabetes through miR-130a-3p/ALK2 Axis. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:6390812. [PMID: 35720932 PMCID: PMC9203195 DOI: 10.1155/2022/6390812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/14/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022]
Abstract
Iron overload is directly associated with diabetes mellitus, loss of islet beta cell, and insulin resistance. Likewise, long noncoding RNA (lncRNA) is associated with type 2 diabetes (T2D). Moreover, lncRNAs could be induced by iron overload. Therefore, we are going to explore the molecular mechanism of lncRNA XIST in iron overload-related T2D. Real-time quantitative PCR and Western blot were used to detect gene and protein levels, respectively. TUNEL and MTT assay were performed to examine cell survival. The glucose test strip, colorimetric analysis kit, ferritin ELISA kit, and insulin ELISA kit were performed to examine the levels of glycolic, iron, and total iron-binding capacity, ferritin, and insulin in serum. Fluorospectrophotometry assay was used to examine labile iron pool level. XIST was higher expressed in T2D and iron overload-related T2D rat tissues and cells, and iron overload-induced promoted XIST expression in T2D. Higher XIST expression was associated with iron overload in patients with T2D. Knockdown of XIST alleviated iron overload and iron overload-induced INS-1 cells injury. Further, we found that XIST can sponge miR-130a-3p to trigger receptor-like kinase 2 (ALK2) expression. Moreover, knockdown of ALK2 alleviated iron overload and iron overload-induced INS-1 cells injury by inhibiting bone morphogenetic protein 6 (BMP6)/ALK2/SMAD1/5/8 axis but reversed with XIST upregulation, which was terminally boosted by overexpression of miR-130a-3p. XIST has the capacity to promote iron overload and iron overload-related T2D initiation and development through inhibition of ALK2 expression by sponging miR-130a-3p, and that targeting this axis may be an effective strategy for treating patients with T2D.
Collapse
Affiliation(s)
- Weiyuan Li
- Department of Geriatrics, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qiu Feng
- Department of Geriatrics, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Chenrong Wang
- Medical Laboratory, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Zhao Yin
- Department of Geriatrics, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xiaolu Li
- Department of Geriatrics, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Lei Li
- Department of Endocrine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
29
|
Karagiannopoulos A, Esguerra JL, Pedersen MG, Wendt A, Prasad RB, Eliasson L. Human pancreatic islet miRNA-mRNA networks of altered miRNAs due to glycemic status. iScience 2022; 25:103995. [PMID: 35310942 PMCID: PMC8927907 DOI: 10.1016/j.isci.2022.103995] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/25/2022] [Accepted: 02/24/2022] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression via mRNA targeting, playing important roles in the pancreatic islets. We aimed to identify molecular pathways and genomic regulatory regions associated with altered miRNA expression due to glycemic status, which could contribute to the development of type 2 diabetes (T2D). To this end, miRNAs were identified by a combination of differential miRNA expression and correlation analysis in human islet samples from donors with normal and elevated blood glucose levels. Analysis and clustering of highly correlated, experimentally validated gene targets of these miRNAs revealed two islet-specific clusters, which were associated with key aspects of islet functions and included a high number of T2D-related genes. Finally, cis-eQTLs and public GWAS data integration uncovered suggestive genomic signals of association with insulin secretion and T2D. The miRNA-driven network-based approach presented in this study contributes to a better understanding of impaired insulin secretion in T2D pathogenesis.
Collapse
Affiliation(s)
- Alexandros Karagiannopoulos
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences-Malmö, Lund University, CRC 91-11, Box 50332, 202 13 Malmö, Sweden
- Clinical Research Centre, Skåne University Hospital, CRC 91-11, Box 50332, 202 13 Malmö, Sweden
| | - Jonathan L.S. Esguerra
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences-Malmö, Lund University, CRC 91-11, Box 50332, 202 13 Malmö, Sweden
- Clinical Research Centre, Skåne University Hospital, CRC 91-11, Box 50332, 202 13 Malmö, Sweden
| | - Morten G. Pedersen
- Department of Information Engineering, University of Padova, Padua, Italy
| | - Anna Wendt
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences-Malmö, Lund University, CRC 91-11, Box 50332, 202 13 Malmö, Sweden
- Clinical Research Centre, Skåne University Hospital, CRC 91-11, Box 50332, 202 13 Malmö, Sweden
| | - Rashmi B. Prasad
- Clinical Research Centre, Skåne University Hospital, CRC 91-11, Box 50332, 202 13 Malmö, Sweden
- Genomics, Diabetes and Endocrinology, Lund University Diabetes Centre Department of Clinical Sciences-Malmö, Lund University, Malmö, Sweden
| | - Lena Eliasson
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences-Malmö, Lund University, CRC 91-11, Box 50332, 202 13 Malmö, Sweden
- Clinical Research Centre, Skåne University Hospital, CRC 91-11, Box 50332, 202 13 Malmö, Sweden
| |
Collapse
|
30
|
San Martín A, Arce-Molina R, Aburto C, Baeza-Lehnert F, Barros LF, Contreras-Baeza Y, Pinilla A, Ruminot I, Rauseo D, Sandoval PY. Visualizing physiological parameters in cells and tissues using genetically encoded indicators for metabolites. Free Radic Biol Med 2022; 182:34-58. [PMID: 35183660 DOI: 10.1016/j.freeradbiomed.2022.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/07/2023]
Abstract
The study of metabolism is undergoing a renaissance. Since the year 2002, over 50 genetically-encoded fluorescent indicators (GEFIs) have been introduced, capable of monitoring metabolites with high spatial/temporal resolution using fluorescence microscopy. Indicators are fusion proteins that change their fluorescence upon binding a specific metabolite. There are indicators for sugars, monocarboxylates, Krebs cycle intermediates, amino acids, cofactors, and energy nucleotides. They permit monitoring relative levels, concentrations, and fluxes in living systems. At a minimum they report relative levels and, in some cases, absolute concentrations may be obtained by performing ad hoc calibration protocols. Proper data collection, processing, and interpretation are critical to take full advantage of these new tools. This review offers a survey of the metabolic indicators that have been validated in mammalian systems. Minimally invasive, these indicators have been instrumental for the purposes of confirmation, rebuttal and discovery. We envision that this powerful technology will foster metabolic physiology.
Collapse
Affiliation(s)
- A San Martín
- Centro de Estudios Científicos (CECs), Valdivia, Chile.
| | - R Arce-Molina
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - C Aburto
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | | | - L F Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - Y Contreras-Baeza
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - A Pinilla
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - I Ruminot
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - D Rauseo
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - P Y Sandoval
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| |
Collapse
|
31
|
Saleh AA, El-Hefnawy SM, Kasemy ZA, Alhagaa AA, Nooh MZ, Arafat ES. Mi-RNA-93 and Mi-RNA-152 in the Diagnosis of Type 2 Diabetes and Diabetic Retinopathy. Br J Biomed Sci 2022; 79:10192. [PMID: 35996507 PMCID: PMC8915732 DOI: 10.3389/bjbs.2021.10192] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/23/2021] [Indexed: 12/03/2022]
Abstract
Background and Aim: Diabetes mellitus (DM) is a chronic disorder with diabetic retinopathy (DR) as one of its main microvascular outcomes, being a prime cause of vision loss. Dysregulation of microRNAs (miRNAs) has been associated with some diabetic microvascular complications such as diabetic retinopathy. This hypothesised changes in the serum of miR-93 and miR-152 in diabetes and diabetic retinopathy. Methods: The study cohort consisted of 80 healthy volunteers, 80 type 2 diabetic patients, and 80 diabetic retinopathy patients, of whom 40 had proliferative (PDR) and 40 non-proliferative retinopathy (NPDR). Serum fasting and 2-hour postprandial glucose (2hPP), glycated haemoglobin (HbA1c), fasting insulin, and HOMA-IR were evaluated by routine methods, miR-93 and miR-152 expression by quantitative real-time PCR. Results: FBG, 2hPP, fasting insulin, HOMA-IR, and miR-152 showed an increasing trend across groups while miR-93 showed a decreasing trend (all p < 0.001). Binary logistic regression analysis for prediction of DR found that the most significant were miR-152 (OR 1.37, 95% CI: 1.18-1.58, <0.001), BMI (1.13, [1.07-1.31], p = 0.004), duration of disease (1.29 [1.04-1.6] p = 0.018), and miR-152 (0.01, [0.0-0.47] p = 0.019). The most significant predictors of PDR were miR-152 (OR = 1.47, 95% CI: 1.12-1.92, p = 0.005), HOMA-IR (2.66 [1.30-5.45] p = 0.007), and miR-93 (0.25 [0.07-0.86] p = 0.028). Conclusion: MiR-93 and miR-152 can differentiate patients with diabetes and those with DR. Both miRNAs might be potential biomarkers for diabetes and diabetic retinopathy, and specifically for proliferative diabetic retinopathy.
Collapse
Affiliation(s)
- A. A. Saleh
- Department of Medical Biochemistry and Molecular Biology , Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| | - S. M. El-Hefnawy
- Department of Medical Biochemistry and Molecular Biology , Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| | - Z. A. Kasemy
- Department of Public Health and Community Medicine, Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| | - A. A. Alhagaa
- Department of Ophthalmology, Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| | - M. Z. Nooh
- Department of Internal Medicine, Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| | - E. S. Arafat
- Department of Medical Biochemistry and Molecular Biology , Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| |
Collapse
|
32
|
Mir FA, Mall R, Iskandarani A, Ullah E, Samra TA, Cyprian F, Parray A, Alkasem M, Abdalhakam I, Farooq F, Abou-Samra AB. Characteristic MicroRNAs Linked to Dysregulated Metabolic Pathways in Qatari Adult Subjects With Obesity and Metabolic Syndrome. Front Endocrinol (Lausanne) 2022; 13:937089. [PMID: 35937842 PMCID: PMC9352892 DOI: 10.3389/fendo.2022.937089] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/24/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Obesity-associated dysglycemia is associated with metabolic disorders. MicroRNAs (miRNAs) are known regulators of metabolic homeostasis. We aimed to assess the relationship of circulating miRNAs with clinical features in obese Qatari individuals. METHODS We analyzed a dataset of 39 age-matched patients that includes 18 subjects with obesity only (OBO) and 21 subjects with obesity and metabolic syndrome (OBM). We measured 754 well-characterized human microRNAs (miRNAs) and identified differentially expressed miRNAs along with their significant associations with clinical markers in these patients. RESULTS A total of 64 miRNAs were differentially expressed between metabolically healthy obese (OBO) versus metabolically unhealthy obese (OBM) patients. Thirteen out of 64 miRNAs significantly correlated with at least one clinical trait of the metabolic syndrome. Six out of the thirteen demonstrated significant association with HbA1c levels; miR-331-3p, miR-452-3p, and miR-485-5p were over-expressed, whereas miR-153-3p, miR-182-5p, and miR-433-3p were under-expressed in the OBM patients with elevated HbA1c levels. We also identified, miR-106b-3p, miR-652-3p, and miR-93-5p that showed a significant association with creatinine; miR-130b-5p, miR-363-3p, and miR-636 were significantly associated with cholesterol, whereas miR-130a-3p was significantly associated with LDL. Additionally, miR-652-3p's differential expression correlated significantly with HDL and creatinine. CONCLUSIONS MicroRNAs associated with metabolic syndrome in obese subjects may have a pathophysiologic role and can serve as markers for obese individuals predisposed to various metabolic diseases like diabetes.
Collapse
Affiliation(s)
- Fayaz Ahmad Mir
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Raghvendra Mall
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University, Doha, Qatar
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Ahmad Iskandarani
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ehsan Ullah
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University, Doha, Qatar
| | - Tareq A Samra
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Farhan Cyprian
- College of Medicine, Qatar University (QU) Health, Qatar University, Doha, Qatar
| | - Aijaz Parray
- Qatar Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Meis Alkasem
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ibrahem Abdalhakam
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Faisal Farooq
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University, Doha, Qatar
| | - Abdul-Badi Abou-Samra
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
33
|
Sałówka A, Martinez-Sanchez A. Molecular Mechanisms of Nutrient-Mediated Regulation of MicroRNAs in Pancreatic β-cells. Front Endocrinol (Lausanne) 2021; 12:704824. [PMID: 34803905 PMCID: PMC8600252 DOI: 10.3389/fendo.2021.704824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic β-cells within the islets of Langerhans respond to rising blood glucose levels by secreting insulin that stimulates glucose uptake by peripheral tissues to maintain whole body energy homeostasis. To different extents, failure of β-cell function and/or β-cell loss contribute to the development of Type 1 and Type 2 diabetes. Chronically elevated glycaemia and high circulating free fatty acids, as often seen in obese diabetics, accelerate β-cell failure and the development of the disease. MiRNAs are essential for endocrine development and for mature pancreatic β-cell function and are dysregulated in diabetes. In this review, we summarize the different molecular mechanisms that control miRNA expression and function, including transcription, stability, posttranscriptional modifications, and interaction with RNA binding proteins and other non-coding RNAs. We also discuss which of these mechanisms are responsible for the nutrient-mediated regulation of the activity of β-cell miRNAs and identify some of the more important knowledge gaps in the field.
Collapse
Affiliation(s)
| | - Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
34
|
Dittmar RL, Liu S, Tai MC, Rajapakshe K, Huang Y, Longton G, DeCapite C, Hurd MW, Paris PL, Kirkwood KS, Coarfa C, Maitra A, Brand RE, Killary AM, Sen S. Plasma miRNA Biomarkers in Limited Volume Samples for Detection of Early-stage Pancreatic Cancer. Cancer Prev Res (Phila) 2021; 14:729-740. [PMID: 33893071 PMCID: PMC8818322 DOI: 10.1158/1940-6207.capr-20-0303] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/11/2020] [Accepted: 04/21/2021] [Indexed: 11/16/2022]
Abstract
Early detection of pancreatic ductal adenocarcinoma (PDAC) is key to improving patient outcomes; however, PDAC is usually diagnosed late. Therefore, blood-based minimally invasive biomarker assays for limited volume clinical samples are urgently needed. A novel miRNA profiling platform (Abcam Fireplex-Oncology Panel) was used to investigate the feasibility of developing early detection miRNA biomarkers with 20 μL plasma from a training set (58 stage II PDAC cases and 30 controls) and two validation sets (34 stage II PDAC cases and 25 controls; 44 stage II PDAC cases and 18 controls). miR-34a-5p [AUC = 0.77; 95% confidence interval (CI), 0.66-0.87], miR-130a-3p (AUC = 0.74; 95% CI, 0.63-0.84), and miR-222-3p (AUC = 0.70; 95% CI, 0.58-0.81) were identified as significantly differentially abundant in plasma from stage II PDAC versus controls. Although none of the miRNAs individually outperformed the currently used serologic biomarker for PDAC, carbohydrate antigen 19-9 (CA19-9), combining the miRNAs with CA 19-9 improved AUCs from 0.89 (95% CI, 0.81-0.95) for CA 19-9 alone to 0.92 (95% CI, 0.86-0.97), 0.94 (95% CI, 0.89-0.98), and 0.92 (95% CI, 0.87-0.97), respectively. Gene set enrichment analyses of transcripts correlated with high and low expression of the three miRNAs in The Cancer Genome Atlas PDAC sample set. These miRNA biomarkers, assayed in limited volume plasma together with CA19-9, discriminate stage II PDAC from controls with good sensitivity and specificity. Unbiased profiling of larger cohorts should help develop an informative early detection biomarker assay for diagnostic settings. PREVENTION RELEVANCE: Development of minimally invasive biomarker assays for detection of premalignant disease and early-stage pancreatic cancer is key to improving patient survival. This study describes a limited volume plasma miRNA biomarker assay that can detect early-stage resectable pancreatic cancer in clinical samples necessary for effective prevention and clinical intervention.
Collapse
Affiliation(s)
- Rachel L Dittmar
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
- University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| | - Suyu Liu
- University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
- Department of Biostatistics, Division of Science, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mei Chee Tai
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Ying Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Biostatistics University of Washington, Seattle, Washington
| | - Gary Longton
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Christine DeCapite
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Mark W Hurd
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pamela L Paris
- Department of Urology and Division of Hematology Oncology, UCSF Helen Diller Cancer Research Center, San Francisco, California
| | - Kimberly S Kirkwood
- Department of Surgery, Division of General Surgery, UCSF Helen Diller Cancer Research Center, San Francisco, California
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Anirban Maitra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
- University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| | - Randall E Brand
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ann M Killary
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
- University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| | - Subrata Sen
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas.
- University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
35
|
MicroRNA-mediated regulation of glucose and lipid metabolism. Nat Rev Mol Cell Biol 2021; 22:425-438. [PMID: 33772227 PMCID: PMC8853826 DOI: 10.1038/s41580-021-00354-w] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2021] [Indexed: 02/01/2023]
Abstract
In animals, systemic control of metabolism is conducted by metabolic tissues and relies on the regulated circulation of a plethora of molecules, such as hormones and lipoprotein complexes. MicroRNAs (miRNAs) are a family of post-transcriptional gene repressors that are present throughout the animal kingdom and have been widely associated with the regulation of gene expression in various contexts, including virtually all aspects of systemic control of metabolism. Here we focus on glucose and lipid metabolism and review current knowledge of the role of miRNAs in their systemic regulation. We survey miRNA-mediated regulation of healthy metabolism as well as the contribution of miRNAs to metabolic dysfunction in disease, particularly diabetes, obesity and liver disease. Although most miRNAs act on the tissue they are produced in, it is now well established that miRNAs can also circulate in bodily fluids, including their intercellular transport by extracellular vesicles, and we discuss the role of such extracellular miRNAs in systemic metabolic control and as potential biomarkers of metabolic status and metabolic disease.
Collapse
|
36
|
Chi T, Lin J, Wang M, Zhao Y, Liao Z, Wei P. Non-Coding RNA as Biomarkers for Type 2 Diabetes Development and Clinical Management. Front Endocrinol (Lausanne) 2021; 12:630032. [PMID: 34603195 PMCID: PMC8484715 DOI: 10.3389/fendo.2021.630032] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 08/10/2021] [Indexed: 12/21/2022] Open
Abstract
Diabetes, a metabolic disease characterized by high blood glucose and other complications, has undefined causes and multiple risk factors, including inappropriate diet, unhealthy lifestyles, and genetic predisposition. The two most distinguished types of diabetes are type 1 and type 2 diabetes, resulting from the autoimmune impairment of insulin-generating pancreatic β cells and insulin insensitivity, respectively. Non-coding RNAs (ncRNAs), a cohort of RNAs with little transcriptional value, have been found to exert substantial importance in epigenetic and posttranscriptional modulation of gene expression such as messenger RNA (mRNA) silencing. This review mainly focuses on the pathology of type 2 diabetes (T2D) and ncRNAs as potential biomarkers in T2D development and clinical management. We consolidate the pathogenesis, diagnosis, and current treatments of T2D, and present the existing evidence on changes in multiple types of ncRNAs in response to various pathological changes and dysfunctions in different stages of T2D.
Collapse
Affiliation(s)
- Tiange Chi
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaran Lin
- Department of Nephrology and Endocrinology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Mina Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, China
| | - Yihan Zhao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
- *Correspondence: Peng Wei, ; Zehuan Liao,
| | - Peng Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Peng Wei, ; Zehuan Liao,
| |
Collapse
|
37
|
Chen YJ, Lin TL, Cai Z, Yan CH, Gou SR, Zhuang YD. Assessment of acute pancreatitis severity via determination of serum levels of hsa-miR-126-5p and IL-6. Exp Ther Med 2020; 21:26. [PMID: 33262812 PMCID: PMC7690249 DOI: 10.3892/etm.2020.9458] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 07/23/2020] [Indexed: 12/14/2022] Open
Abstract
Early assessment of acute pancreatitis (AP) severity is key to its treatment. The present study aimed to explore the role of microRNAs (miRNAs/miRs) combined with inflammatory factors in determining AP severity. For this, serum pro-inflammatory cytokines [tumor necrosis factor (TNF)-α, interleukin (IL)-1, IL-6, IL-8 and IL-10)] and miRNAs [Homo sapiens (hsa)-miR-548d-5p, hsa-miR-126-5p and hsa-miR-130b-5p] were detected in patients with mild AP (MAP), severe AP (SAP) and recurrent AP (RAP). High expression of IL-10, TNF-α, hsa-miR-126-5p, hsa-miR-548d-5p and hsa-miR-130b-5p was able to distinguish SAP from MAP and RAP (P<0.05). Multifactorial binary logistic regression analysis indicated that IL-1/IL-6 combined with hsa-miR-126-5p/hsa-miR-548d-5p had a significant influence on AP and AP severity (P<0.05). Receiver operating characteristic analysis revealed that IL-1 combined with hsa-miR-126-5p [area under the curve (AUC), 0.926; sensitivity, 90.0%; specificity, 86.7%, P<0.001] and IL-6 combined with hsa-miR-126-5p (AUC, 0.952; sensitivity, 93.3%; specificity, 90.0%; P<0.001) were able to better distinguish MAP from SAP than IL-1/IL-6 combined with hsa-miR-548d-5p, lipase, and amylase. IL-1 or IL-6 combined with hsa-miR-548d-5p (AUC, 0.924; sensitivity, 83.3%; specificity, 93.3%; P<0.001) were able to better distinguish SAP from RAP than IL-1/IL-6 combined with hsa-miR-126-5p, lipase, and amylase. IL-1 combined with hsa-miR-126-5p (AUC, 0.926; sensitivity, 90.0%; specificity, 86.7%; P<0.001) and IL-6 combined with hsa-miR-126-5p (AUC, 0.952; sensitivity, 93.3%; specificity, 90.0%; P<0.001) were able to better differentiate between MAP and RAP than IL-1/IL-6 combined with hsa-miR-548d-5p, lipase, and amylase. These results demonstrated that the combined detection of serum IL-6 and hsa-miR-126-5p may be useful for the early prediction of AP classification.
Collapse
Affiliation(s)
- Ying-Jie Chen
- Department of Critical Care Medicine, Jinjiang Hospital of Traditional Chinese Medicine, Jinjiang, Fujian 362200, P.R. China
| | - Tian-Lai Lin
- Department of Critical Care Medicine, Quanzhou First Hospital Affiliated to Fujian Medicine University, Quanzhou, Fujian 362000, P.R. China
| | - Zhe Cai
- Department of Emergency Medicine, Jinjiang Hospital of Traditional Chinese Medicine, Jinjiang, Fujian 362200, P.R. China
| | - Chang-Hu Yan
- Department of Critical Care Medicine, Jinjiang Hospital of Traditional Chinese Medicine, Jinjiang, Fujian 362200, P.R. China
| | - Sen-Ren Gou
- Department of Critical Care Medicine, Jinjiang Hospital of Traditional Chinese Medicine, Jinjiang, Fujian 362200, P.R. China
| | - Yao-Dong Zhuang
- Department of Critical Care Medicine, Jinjiang Hospital of Traditional Chinese Medicine, Jinjiang, Fujian 362200, P.R. China
| |
Collapse
|
38
|
Giglio RV, Nikolic D, Volti GL, Stoian AP, Banerjee Y, Magan-Fernandez A, Castellino G, Patti AM, Chianetta R, Castracani CC, Montalto G, Rizvi AA, Sesti G, Rizzo M. Liraglutide Increases Serum Levels of MicroRNA-27b, -130a and -210 in Patients with Type 2 Diabetes Mellitus: A Novel Epigenetic Effect. Metabolites 2020; 10:metabo10100391. [PMID: 33008044 PMCID: PMC7599907 DOI: 10.3390/metabo10100391] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Liraglutide has shown favourable effects on several cardiometabolic risk factors, beyond glucose control. MicroRNAs (miRNAs) regulate gene expression, resulting in post-transcriptional modifications of cell response and function. Specific miRNAs, including miRNA-27b, miRNA-130a, and miRNA-210, play a role in cardiometabolic disease. We aimed to determine the effect of liraglutide on the serum levels of miRNA-27b, miRNA-130a and miRNA-210. Twenty-five subjects with type-2 diabetes mellitus (T2DM), naïve to incretin-based therapy, were treated with liraglutide (1.2 mg/day as an add-on to metformin) for 4 months. miRNAs were quantified using real-time polymerase chain reaction. After liraglutide treatment, we found significant reductions in fasting glucose (from 9.8 ± 5.3 to 6.7 ± 1.6 mmol/L, p = 0.0042), glycosylated haemoglobin (HbA1c) (from 8.1 ± 0.8 to 6.6 ± 1.0%, p = 0.0008), total cholesterol (from 5.0 ± 1.0 to 4.0 ± 0.7 mmol/L, p = 0.0011), triglycerides (from 1.9 ± 1.0 to 1.5 ± 0.8 mmol/L, p = 0.0104) and low-density lipoprotein cholesterol (from 2.9 ± 1.2 to 2.2 ± 0.6 mmol/L, p = 0.0125), while the serum levels of miRNA-27b, miRNA-130a and miRNA-210a were significantly increased (median (interquartile range, IQR) changes: 1.73 (7.12) (p = 0.0401), 1.91 (3.64) (p = 0.0401) and 2.09 (11.0) (p = 0.0486), respectively). Since the changes in miRNAs were independent of changes in all the metabolic parameters investigated, liraglutide seems to exert a direct epigenetic effect in T2DM patients, regulating microRNAs involved in the maintenance of endothelial cell homeostasis. These changes might be implicated in liraglutide’s benefits and may represent useful targets for cardiometabolic management.
Collapse
Affiliation(s)
- Rosaria Vincenza Giglio
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (D.N.); (A.M.-F.); (G.C.); (A.M.P.); (R.C.); (G.M.); (M.R.)
| | - Dragana Nikolic
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (D.N.); (A.M.-F.); (G.C.); (A.M.P.); (R.C.); (G.M.); (M.R.)
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (G.L.V.); (C.C.C.)
| | - Anca Pantea Stoian
- Department of Diabetes, Nutrition and Metabolic Diseases, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Yajnavalka Banerjee
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE;
| | - Antonio Magan-Fernandez
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (D.N.); (A.M.-F.); (G.C.); (A.M.P.); (R.C.); (G.M.); (M.R.)
| | - Giuseppa Castellino
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (D.N.); (A.M.-F.); (G.C.); (A.M.P.); (R.C.); (G.M.); (M.R.)
| | - Angelo Maria Patti
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (D.N.); (A.M.-F.); (G.C.); (A.M.P.); (R.C.); (G.M.); (M.R.)
| | - Roberta Chianetta
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (D.N.); (A.M.-F.); (G.C.); (A.M.P.); (R.C.); (G.M.); (M.R.)
| | - Carlo Castruccio Castracani
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (G.L.V.); (C.C.C.)
| | - Giuseppe Montalto
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (D.N.); (A.M.-F.); (G.C.); (A.M.P.); (R.C.); (G.M.); (M.R.)
| | - Ali A. Rizvi
- Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine, Columbia, SC 29203, USA
- Division of Endocrinology, Metabolism, and Lipids Emory University School of Medicine, Atlanta, GA 30322, USA
- Correspondence: ; Tel.: +1-(404)-778-2064
| | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, University of Rome La Sapienza, 00182 Rome, Italy;
| | - Manfredi Rizzo
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (D.N.); (A.M.-F.); (G.C.); (A.M.P.); (R.C.); (G.M.); (M.R.)
- Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine, Columbia, SC 29203, USA
| |
Collapse
|
39
|
Pathophysiology of Type 2 Diabetes Mellitus. Int J Mol Sci 2020; 21:ijms21176275. [PMID: 32872570 PMCID: PMC7503727 DOI: 10.3390/ijms21176275] [Citation(s) in RCA: 1300] [Impact Index Per Article: 260.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
Type 2 Diabetes Mellitus (T2DM), one of the most common metabolic disorders, is caused by a combination of two primary factors: defective insulin secretion by pancreatic β-cells and the inability of insulin-sensitive tissues to respond appropriately to insulin. Because insulin release and activity are essential processes for glucose homeostasis, the molecular mechanisms involved in the synthesis and release of insulin, as well as in its detection are tightly regulated. Defects in any of the mechanisms involved in these processes can lead to a metabolic imbalance responsible for the development of the disease. This review analyzes the key aspects of T2DM, as well as the molecular mechanisms and pathways implicated in insulin metabolism leading to T2DM and insulin resistance. For that purpose, we summarize the data gathered up until now, focusing especially on insulin synthesis, insulin release, insulin sensing and on the downstream effects on individual insulin-sensitive organs. The review also covers the pathological conditions perpetuating T2DM such as nutritional factors, physical activity, gut dysbiosis and metabolic memory. Additionally, because T2DM is associated with accelerated atherosclerosis development, we review here some of the molecular mechanisms that link T2DM and insulin resistance (IR) as well as cardiovascular risk as one of the most important complications in T2DM.
Collapse
|
40
|
Huey J, Keutler K, Schultz C. Chemical Biology Toolbox for Studying Pancreatic Islet Function - A Perspective. Cell Chem Biol 2020; 27:1015-1031. [PMID: 32822616 DOI: 10.1016/j.chembiol.2020.07.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/10/2020] [Accepted: 07/28/2020] [Indexed: 01/14/2023]
Abstract
The islets of Langerhans represent one of the many complex endocrine organs in mammals. Traditionally, islet function is studied by a mixture of physiological, cell biological, and molecular biological methods. Recently, novel techniques stemming from the ever-increasing toolbox provided by chemical laboratories have been added to the repertoire. Many emerging techniques will soon be available to manipulate and monitor islet function at the single-cell level and potentially in intact model animals, as well as in isolated human islets. Here, we review the most current small-molecule-based and genetically encoded molecular tool sets available to study islet function. We provide an outlook regarding future tool developments that will impact islet research, with a special focus on the interplay between different islet cell types.
Collapse
Affiliation(s)
- Julia Huey
- Program in Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97210, USA; Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97210, USA
| | - Kaya Keutler
- Program in Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97210, USA; Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97210, USA
| | - Carsten Schultz
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97210, USA.
| |
Collapse
|
41
|
Amouyal C, Castel J, Guay C, Lacombe A, Denom J, Migrenne-Li S, Rouault C, Marquet F, Georgiadou E, Stylianides T, Luquet S, Le Stunff H, Scharfmann R, Clément K, Rutter GA, Taboureau O, Magnan C, Regazzi R, Andreelli F. A surrogate of Roux-en-Y gastric bypass (the enterogastro anastomosis surgery) regulates multiple beta-cell pathways during resolution of diabetes in ob/ob mice. EBioMedicine 2020; 58:102895. [PMID: 32739864 PMCID: PMC7393530 DOI: 10.1016/j.ebiom.2020.102895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bariatric surgery is an effective treatment for type 2 diabetes. Early post-surgical enhancement of insulin secretion is key for diabetes remission. The full complement of mechanisms responsible for improved pancreatic beta cell functionality after bariatric surgery is still unclear. Our aim was to identify pathways, evident in the islet transcriptome, that characterize the adaptive response to bariatric surgery independently of body weight changes. METHODS We performed entero-gastro-anastomosis (EGA) with pyloric ligature in leptin-deficient ob/ob mice as a surrogate of Roux-en-Y gastric bypass (RYGB) in humans. Multiple approaches such as determination of glucose tolerance, GLP-1 and insulin secretion, whole body insulin sensitivity, ex vivo glucose-stimulated insulin secretion (GSIS) and functional multicellular Ca2+-imaging, profiling of mRNA and of miRNA expression were utilized to identify significant biological processes involved in pancreatic islet recovery. FINDINGS EGA resolved diabetes, increased pancreatic insulin content and GSIS despite a persistent increase in fat mass, systemic and intra-islet inflammation, and lipotoxicity. Surgery differentially regulated 193 genes in the islet, most of which were involved in the regulation of glucose metabolism, insulin secretion, calcium signaling or beta cell viability, and these were normalized alongside changes in glucose metabolism, intracellular Ca2+ dynamics and the threshold for GSIS. Furthermore, 27 islet miRNAs were differentially regulated, four of them hubs in a miRNA-gene interaction network and four others part of a blood signature of diabetes resolution in ob/ob mice and in humans. INTERPRETATION Taken together, our data highlight novel miRNA-gene interactions in the pancreatic islet during the resolution of diabetes after bariatric surgery that form part of a blood signature of diabetes reversal. FUNDING European Union's Horizon 2020 research and innovation programme via the Innovative Medicines Initiative 2 Joint Undertaking (RHAPSODY), INSERM, Société Francophone du Diabète, Institut Benjamin Delessert, Wellcome Trust Investigator Award (212625/Z/18/Z), MRC Programme grants (MR/R022259/1, MR/J0003042/1, MR/L020149/1), Diabetes UK (BDA/11/0004210, BDA/15/0005275, BDA 16/0005485) project grants, National Science Foundation (310030-188447), Fondation de l'Avenir.
Collapse
Affiliation(s)
- Chloé Amouyal
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic approaches (NutriOmics), Paris, France; AP-HP, Pitié-Salpêtrière Hospital, Diabetology department, F-75013 Paris, France
| | - Julien Castel
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Claudiane Guay
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, CH-1005, Lausanne, Switzerland
| | - Amélie Lacombe
- PreclinICAN, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Jessica Denom
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | | | - Christine Rouault
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic approaches (NutriOmics), Paris, France
| | - Florian Marquet
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic approaches (NutriOmics), Paris, France
| | - Eleni Georgiadou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | | - Serge Luquet
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Hervé Le Stunff
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Raphael Scharfmann
- Université de Paris, Cochin Institute, Inserm U1016, Paris 75014, France
| | - Karine Clément
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic approaches (NutriOmics), Paris, France; APHP, Pitié-Salpêtrière Hospital, Nutrition department, F-75013 Paris, France
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK; Lee Kong Chian School of Medicine, Nan Yang Technological University, Singapore
| | - Olivier Taboureau
- Université de Paris, BFA, Team CMPLI, Inserm U1133, CNRS UMR 8251, Paris, France
| | | | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, CH-1005, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Rue du Bugnon 7, CH-1005 Lausanne, Switzerland
| | - Fabrizio Andreelli
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic approaches (NutriOmics), Paris, France; AP-HP, Pitié-Salpêtrière Hospital, Diabetology department, F-75013 Paris, France.
| |
Collapse
|
42
|
Guay C, Jacovetti C, Bayazit MB, Brozzi F, Rodriguez-Trejo A, Wu K, Regazzi R. Roles of Noncoding RNAs in Islet Biology. Compr Physiol 2020; 10:893-932. [PMID: 32941685 DOI: 10.1002/cphy.c190032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The discovery that most mammalian genome sequences are transcribed to ribonucleic acids (RNA) has revolutionized our understanding of the mechanisms governing key cellular processes and of the causes of human diseases, including diabetes mellitus. Pancreatic islet cells were found to contain thousands of noncoding RNAs (ncRNAs), including micro-RNAs (miRNAs), PIWI-associated RNAs, small nucleolar RNAs, tRNA-derived fragments, long non-coding RNAs, and circular RNAs. While the involvement of miRNAs in islet function and in the etiology of diabetes is now well documented, there is emerging evidence indicating that other classes of ncRNAs are also participating in different aspects of islet physiology. The aim of this article will be to provide a comprehensive and updated view of the studies carried out in human samples and rodent models over the past 15 years on the role of ncRNAs in the control of α- and β-cell development and function and to highlight the recent discoveries in the field. We not only describe the role of ncRNAs in the control of insulin and glucagon secretion but also address the contribution of these regulatory molecules in the proliferation and survival of islet cells under physiological and pathological conditions. It is now well established that most cells release part of their ncRNAs inside small extracellular vesicles, allowing the delivery of genetic material to neighboring or distantly located target cells. The role of these secreted RNAs in cell-to-cell communication between β-cells and other metabolic tissues as well as their potential use as diabetes biomarkers will be discussed. © 2020 American Physiological Society. Compr Physiol 10:893-932, 2020.
Collapse
Affiliation(s)
- Claudiane Guay
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Cécile Jacovetti
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Mustafa Bilal Bayazit
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Flora Brozzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Adriana Rodriguez-Trejo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Kejing Wu
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
43
|
MicroRNA-130a targeting hypoxia-inducible factor 1 alpha suppresses cell metastasis and Warburg effect of NSCLC cells under hypoxia. Life Sci 2020; 255:117826. [PMID: 32450163 DOI: 10.1016/j.lfs.2020.117826] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022]
Abstract
MicroRNAs have been demonstrated to play critical role in the development of non-small cell lung cancer (NSCLC) and hypoxia is a common hallmark of NSCLC. MiRNA-130a-3p (miR-130a) is a well-known tumor suppressor, and we intended to explore the role and mechanism of miR-130a in NSCLC cells under hypoxia. We used real-time quantitative polymerase chain reaction method to measure miR-130a expression, and found that miR-130a was downregulated in human NSCLC tumors and cell lines (A549 and H1299), accompanied with upregulation of hypoxia-inducible factor 1 alpha (HIF1A), a marker of hypoxia. Besides, miR-130a low expression was associated with tumor burden and poor overall survival. Moreover, miR-130a expression was even downregulated in hypoxia-treated A549 and H1299 cells. Ectopic expression of miR-130a suppressed Warburg effect, migration and invasion in hypoxic A549 and H1299 cells, as evidenced by decreased glucose consumption, lactate production, hexokinase 2 expression, and numbers of migration cells and invasion cells analyzed by commercial glucose and lactate assay kits, western blotting and transwell assays. Furthermore, overexpression of miR-130a restrained xenograft tumor growth of A549 cells in mice. However, recovery of HIF1A could reverse the suppressive effect of miR-130a overexpression on cell migration, invasion and Warburg effect in hypoxic A549 and H1299 cells. Mechanically, dual-luciferase reporter assay, RNA immunoprecipitation and RNA pull-down assay confirmed a target relationship between miR-130a and HIF1A. Collectively, we demonstrated an anti-tumor role of miR-130a in NSCLC cells under hypoxia through targeting HIF1A, suggesting a potential target for the interfering of NSCLC.
Collapse
|
44
|
Das S, Mohamed IN, Teoh SL, Thevaraj T, Ku Ahmad Nasir KN, Zawawi A, Salim HH, Zhou DK. Micro-RNA and the Features of Metabolic Syndrome: A Narrative Review. Mini Rev Med Chem 2020; 20:626-635. [DOI: 10.2174/1389557520666200122124445] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/30/2019] [Accepted: 01/04/2020] [Indexed: 12/19/2022]
Abstract
The incidence of Metabolic Syndrome (MetS) has risen globally. MetS includes a combination
of features, i.e. blood glucose impairment, excess abdominal/body fat dyslipidemia and elevated
blood pressure. Other than conventional treatment with drugs, the main preventive approaches include
lifestyle changes, weight loss, diet control and adequate exercise also proves to be beneficial. MicroRNAs
(miRNAs) are small non-coding RNAs that play critical regulatory roles in most biological
and pathological processes. In the present review, we discuss various miRNAs which are related to
MetS by targeting various organs, including the pancreas, liver, skeletal muscles and adipose tissues.
These miRNAs have the effect on insulin production and secretion (miR-9, miR-124a, miR-130a,b,
miR152, miR-335, miR-375), insulin resistance (miR-29), adipogenesis (miR-143, miR148a) and lipid
metabolism (miR-192). We also discuss the miRNAs as potential biomarkers and future therapeutic
targets. This review may be beneficial for molecular biologists and clinicians dealing with MetS.
Collapse
Affiliation(s)
- Srijit Das
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Isa Naina Mohamed
- Department of Pharmacology, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Tarrsini Thevaraj
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | | | - Azwani Zawawi
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Hazwan Hazrin Salim
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Dennis Kheng Zhou
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
45
|
Eliasson L, Esguerra JLS. MicroRNA Networks in Pancreatic Islet Cells: Normal Function and Type 2 Diabetes. Diabetes 2020; 69:804-812. [PMID: 32312896 PMCID: PMC7171954 DOI: 10.2337/dbi19-0016] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Abstract
Impaired insulin secretion from the pancreatic β-cells is central in the pathogenesis of type 2 diabetes (T2D), and microRNAs (miRNAs) are fundamental regulatory factors in this process. Differential expression of miRNAs contributes to β-cell adaptation to compensate for increased insulin resistance, but deregulation of miRNA expression can also directly cause β-cell impairment during the development of T2D. miRNAs are small noncoding RNAs that posttranscriptionally reduce gene expression through translational inhibition or mRNA destabilization. The nature of miRNA targeting implies the presence of complex and large miRNA-mRNA regulatory networks in every cell, including the insulin-secreting β-cell. Here we exemplify one such network using our own data on differential miRNA expression in the islets of T2D Goto-Kakizaki rat model. Several biological processes are influenced by multiple miRNAs in the β-cell, but so far most studies have focused on dissecting the mechanism of action of individual miRNAs. In this Perspective we present key islet miRNA families involved in T2D pathogenesis including miR-200, miR-7, miR-184, miR-212/miR-132, and miR-130a/b/miR-152. Finally, we highlight four challenges and opportunities within islet miRNA research, ending with a discussion on how miRNAs can be utilized as therapeutic targets contributing to personalized T2D treatment strategies.
Collapse
Affiliation(s)
- Lena Eliasson
- Islet Cell Exocytosis, Lund University Diabetes Centre; Department of Clinical Sciences Malmö, Lund University; and Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Jonathan L S Esguerra
- Islet Cell Exocytosis, Lund University Diabetes Centre; Department of Clinical Sciences Malmö, Lund University; and Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
46
|
Micro(RNA) Management and Mismanagement of the Islet. J Mol Biol 2020; 432:1419-1428. [DOI: 10.1016/j.jmb.2019.09.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/10/2019] [Accepted: 09/15/2019] [Indexed: 02/08/2023]
|
47
|
Zhang L, Wu H, Zhao M, Lu Q. Identifying the differentially expressed microRNAs in autoimmunity: A systemic review and meta-analysis. Autoimmunity 2020; 53:122-136. [DOI: 10.1080/08916934.2019.1710135] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Lian Zhang
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Central South University, Changsha, China
| | - Haijing Wu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Central South University, Changsha, China
| | - Ming Zhao
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Central South University, Changsha, China
| | - Qianjin Lu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Central South University, Changsha, China
| |
Collapse
|
48
|
Nagao M, Esguerra JLS, Wendt A, Asai A, Sugihara H, Oikawa S, Eliasson L. Selectively Bred Diabetes Models: GK Rats, NSY Mice, and ON Mice. Methods Mol Biol 2020; 2128:25-54. [PMID: 32180184 DOI: 10.1007/978-1-0716-0385-7_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The polygenic background of selectively bred diabetes models mimics the etiology of type 2 diabetes. So far, three different rodent models (Goto-Kakizaki rats, Nagoya-Shibata-Yasuda mice, and Oikawa-Nagao mice) have been established in the diabetes research field by continuous selective breeding for glucose tolerance from outbred rodent stocks. The origin of hyperglycemia in these rodents is mainly insulin secretion deficiency from the pancreatic β-cells and mild insulin resistance in insulin target organs. In this chapter, we summarize backgrounds and phenotypes of these rodent models to highlight their importance in diabetes research. Then, we introduce experimental methodologies to evaluate β-cell exocytosis as a putative common defect observed in these rodent models.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/etiology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/etiology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Exocytosis
- Gene Expression Profiling/methods
- Glucose Intolerance
- Insulin Resistance/physiology
- Insulin Secretion/physiology
- Insulin-Secreting Cells/chemistry
- Insulin-Secreting Cells/cytology
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/physiology
- Mice
- Mice, Inbred C3H
- Patch-Clamp Techniques/methods
- Phenotype
- Rats
- Rats, Wistar
- Selective Breeding/genetics
Collapse
Affiliation(s)
- Mototsugu Nagao
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.
- Clinical Research Centre, Skåne University Hospital, Lund and Malmö, Sweden.
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.
| | - Jonathan Lou S Esguerra
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Clinical Research Centre, Skåne University Hospital, Lund and Malmö, Sweden
| | - Anna Wendt
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Clinical Research Centre, Skåne University Hospital, Lund and Malmö, Sweden
| | - Akira Asai
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Food and Health Science Research Unit, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Hitoshi Sugihara
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Shinichi Oikawa
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Diabetes and Lifestyle-related Disease Center, Japan Anti-Tuberculosis Association, Fukujuji Hospital, Tokyo, Japan
| | - Lena Eliasson
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.
- Clinical Research Centre, Skåne University Hospital, Lund and Malmö, Sweden.
| |
Collapse
|
49
|
Weighted Gene Coexpression Network Analysis Identified MicroRNA Coexpression Modules and Related Pathways in Type 2 Diabetes Mellitus. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9567641. [PMID: 31915515 PMCID: PMC6935443 DOI: 10.1155/2019/9567641] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/04/2019] [Accepted: 11/11/2019] [Indexed: 12/31/2022]
Abstract
Objective Type 2 diabetes mellitus (T2DM) is a metabolic disease with high incidence, which has seriously affected human life and health. MicroRNA, a short-chain noncoding RNA, plays an important role in T2DM. Identification of meaningful microRNA modules and the role of microRNAs provide a basis for searching potential biomarkers of T2DM. Materials and Methods In this study, three newly diagnosed patients with T2DM and three controls were selected for Whole Peripheral Blood RNA Sequencing to establish a microRNA library. Weighted gene coexpression network analysis (WGCNA) was applied to construct coexpression modules and to detect the trait-related microRNA modules; then, KEGG enrichment analysis was performed to predict the biological function of the interest modules, and candidate hub microRNAs were screened out by the value of module membership (MM) and protein-protein interaction (PPI) network. Result Four microRNA modules (blue, brown, magenta, and turquoise) were highly associated with the T2DM; the number of miRNAs in these modules ranged from 41 to 469. The Fc gamma R-mediated phagocytosis pathway, Rap1 signaling pathway, MAPK signaling pathway, and Lysosome pathway were common pathways in three of the four modules. RPS27A, UBC, and RAC1 were the top three proteins in our study; their corresponding RNAs were miR-1271-5p, miR-130a-3p, miR-130b-3p, and miR-574-3p. Conclusion In summary, this study identified blood miRNAs in human T2DM using RNA sequencing. The findings may be the foundation for understanding the potential role of miRNAs in T2DM.
Collapse
|
50
|
Melnik BC. Milk exosomal miRNAs: potential drivers of AMPK-to-mTORC1 switching in β-cell de-differentiation of type 2 diabetes mellitus. Nutr Metab (Lond) 2019; 16:85. [PMID: 31827573 PMCID: PMC6898964 DOI: 10.1186/s12986-019-0412-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/22/2019] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) steadily increases in prevalence since the 1950's, the period of widespread distribution of refrigerated pasteurized cow's milk. Whereas breastfeeding protects against the development of T2DM in later life, accumulating epidemiological evidence underlines the role of cow's milk consumption in T2DM. Recent studies in rodent models demonstrate that during the breastfeeding period pancreatic β-cells are metabolically immature and preferentially proliferate by activation of mechanistic target of rapamycin complex 1 (mTORC1) and suppression of AMP-activated protein kinase (AMPK). Weaning determines a metabolic switch of β-cells from a proliferating, immature phenotype with low insulin secretion to a differentiated mature phenotype with glucose-stimulated insulin secretion, less proliferation, reduced mTORC1- but increased AMPK activity. Translational evidence presented in this perspective implies for the first time that termination of milk miRNA transfer is the driver of this metabolic switch. miRNA-148a is a key inhibitor of AMPK and phosphatase and tensin homolog, crucial suppressors of mTORC1. β-Cells of diabetic patients return to the postnatal phenotype with high mTORC1 and low AMPK activity, explained by continuous transfer of bovine milk miRNAs to the human milk consumer. Bovine milk miRNA-148a apparently promotes β-cell de-differentiation to the immature mTORC1-high/AMPK-low phenotype with functional impairments in insulin secretion, increased mTORC1-driven endoplasmic reticulum stress, reduced autophagy and early β-cell apoptosis. In contrast to pasteurized cow's milk, milk's miRNAs are inactivated by bacterial fermentation, boiling and ultra-heat treatment and are missing in current infant formula. Persistent milk miRNA signaling adds a new perspective to the pathogenesis of T2DM and explains the protective role of breastfeeding but the diabetogenic effect of continued milk miRNA signaling by persistent consumption of pasteurized cow's milk.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Am Finkenhügel 7A, D-49076 Osnabrück, Germany
| |
Collapse
|