1
|
Cho H, Ha SE, Singh R, Kim D, Ro S. microRNAs in Type 1 Diabetes: Roles, Pathological Mechanisms, and Therapeutic Potential. Int J Mol Sci 2025; 26:3301. [PMID: 40244147 PMCID: PMC11990060 DOI: 10.3390/ijms26073301] [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: 02/21/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by the progressive destruction of pancreatic β-cells, leading to insulin deficiency. The primary drivers of β-cell destruction in T1D involve autoimmune-mediated processes that trigger chronic inflammation and ultimately β-cell loss. Regulatory microRNAs (miRNAs) play a crucial role in modulating these processes by regulating gene expression through post-transcriptional suppression of target mRNAs. Dysregulated miRNAs have been implicated in T1D pathogenesis, serving as both potential diagnostic biomarkers and therapeutic targets. This review explores the role of miRNAs in T1D, highlighting their involvement in disease mechanisms across both rodent models and human patients. While current antidiabetic therapies manage T1D symptoms, they do not prevent β-cell destruction, leaving patients reliant on lifelong insulin therapy. By summarizing key miRNA expression profiles in diabetic animal models and patients, this review explores the potential of miRNA-based therapies to restore β-cell function and halt or slow the progression of the disease.
Collapse
Affiliation(s)
| | | | | | | | - Seungil Ro
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (H.C.); (S.E.H.); (R.S.); (D.K.)
| |
Collapse
|
2
|
Kuang YL, Locatelli CA, Qin Y, Zhang Y, Theusch E, Muñoz-Howell A, Sanchez G, Lu M, Nguyen MA, Yalamanchili T, Wang X, Nalula G, Mattis AN, Oni-Orisan A, Iribarren C, Krauss RM, Mulvihill EE, Medina MW. MIR192 Upregulates GLP-1 Receptor and Improves Statin-Induced Impairment of Insulin Secretion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643960. [PMID: 40166140 PMCID: PMC11956930 DOI: 10.1101/2025.03.18.643960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Statins are a commonly prescribed cholesterol lowering drug class that can increase the risk of new-onset diabetes (NOD). To investigate the molecular mechanisms underlying this effect, we generated human induced pluripotent stem cells (iPSCs) from individuals identified from electronic health records of Kaiser Permanente of Northern California who were susceptible to developing NOD after statin initiation or controls who maintained stable fasting glucose on statin treatment. RNA-seq analysis of iPSCs incubated with atorvastatin, simvastatin or mock buffer for 24 hours identified the long non-coding RNA MIR194-2HG as a top candidate gene. Statin-induced increases in its expression were observed in NOD resistant controls, while statin-induced reductions occurred in NOD susceptible cases. MIR194-2HG encompasses two microRNA genes: MIR192 and MIR194-2. The mature microRNA miR-192-5p, derived from the 5' arm of MIR192, was predicted to bind the 3'UTR of the glucagon like peptide 1 (GLP-1) receptor (GLP1R) transcript. Transfection of a rat insulinoma cell line INS-1 with a miR-192-5p mimic increased Glp1r transcript (1.41-fold) and protein (1.51-fold) levels compared to a scrambled control. Using a luciferase reporter containing the human GLP1R 3'UTR, miR-192-5p overexpression similarly increased luciferase signal (1.44-fold). The miR-192-5p mimic enhanced glucose stimulated insulin secretion (GSIS) in response to GLP1R agonists (1.64-1.81-fold) and rescued simvastatin-induced GSIS impairment in INS-1 cells. Wildtype mice treated with miR-192 AAV8 had improved glucose sensitivity. Islets isolated from these mice exhibited enhanced GLP-1 potentiated GSIS during perifusion ex vivo. These effects were absent in the DIRKO (Glp1r/Gipr double knockout) mouse islets, consistent with the idea that miR-192 promotes GLP-1 mediated GSIS through GLP1R. These findings implicate MIR192 in statin-induced impairment of GSIS by modulating GLP1R, potentially contributing to the susceptibility to NOD in statin users.
Collapse
Affiliation(s)
- Yu-Lin Kuang
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Cassandra A.A. Locatelli
- Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Yuanyuan Qin
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Yuqing Zhang
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Elizabeth Theusch
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Antonio Muñoz-Howell
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Gabriela Sanchez
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, USA
| | - Meng Lu
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, USA
| | - My-Anh Nguyen
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Tanvi Yalamanchili
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Xuanwen Wang
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Gilbert Nalula
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Aras N. Mattis
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Board Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- The Liver Center, University of California San Francisco, San Francisco, CA, USA
| | - Akinyemi Oni-Orisan
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Carlos Iribarren
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, USA
| | - Ronald M. Krauss
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
- Department of Medicine, University of California San Francisco, Oakland, CA, USA
| | - Erin E. Mulvihill
- Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Marisa W. Medina
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
- The Liver Center, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
3
|
Liu MX, Zhang HF, Liu T, Liu JH, Zhang LQ, Zhu JZ. Abnormal expression of miR-454-3p in type 2 diabetes mellitus induces dysfunction of pancreatic β cells by regulating Yy1. Diab Vasc Dis Res 2025; 22:14791641251335923. [PMID: 40245279 PMCID: PMC12033466 DOI: 10.1177/14791641251335923] [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: 11/20/2024] [Revised: 02/25/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
ObjectiveImpairment of pancreatic β cells is a pathophysiological feature of type 2 diabetes mellitus (T2DM). However, whether abnormally dysregulated miR-454-3p in T2DM is related to the dysfunction of pancreatic β cell remains to be further investigated.MethodsFirst, T2DM patients and healthy subjects were recruited to measure miR-454-3p. Subsequently, pancreatic β cells were cultured with high glucose. The role of miR-454-3p in insulin synthesis, secretion, cell proliferation, and apoptosis were investigated by RT-qPCR, Glucose-stimulated insulin secretion determination, cell counting kit-8, and flow cytometry assays. The target mRNA of miR-454-3p was predicted using bioinformatics software. Then, the targeted binding relationships between the above two factors were verified through RNA Immunoprecipitation and Dual-Luciferase Reporter assays.ResultsThe expression of miR-454-3p was increased in T2DM patients and pancreatic β cells cultured with high glucose. Moreover, miR-454-3p was positively correlated with FPG and HbA1c levels in patients. In cell experiments, miR-454-3p inhibitors significantly improved the function of pancreatic β cells, including increased insulin synthesis and secretion, and promoted proliferation. Moreover, silencing Yy1 reversed the protective effect of miR-454-3p inhibitors on pancreatic β cells.ConclusionmiR-454-3p, which is dysregulated in T2DM, promotes the damage of pancreatic β cells by regulating Yy1, thus aggravating T2DM.
Collapse
Affiliation(s)
- Mei-xiao Liu
- Department of Basic Medicine, Cangzhou Medical College, China
| | - Hai-feng Zhang
- Department of Basic Medicine, Cangzhou Medical College, China
| | - Ting Liu
- Department of Diabetes, Cangzhou Hospital of Integrated TCM-WM, China
| | - Jian-hui Liu
- Department of Basic Medicine, Cangzhou Medical College, China
| | - Lin-qi Zhang
- Department of Chinese Medicine, Xingtai Medical College, Cangzhou, China
| | - Jian-zhong Zhu
- Department of Basic Medicine, Cangzhou Medical College, China
| |
Collapse
|
4
|
Li W, Xu G, Chai GW, Ball A, Zhang Q, Kutryk MJB. The MiR-139-5p and CXCR4 axis may play a role in high glucose-induced inflammation by regulating monocyte migration. Sci Rep 2025; 15:6738. [PMID: 40000897 PMCID: PMC11861593 DOI: 10.1038/s41598-025-91100-1] [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] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
MicroRNAs, a class of small non-coding RNA molecules that regulate gene expression post-transcriptionally, are implicated in various pathological conditions including diabetes mellitus (DM). DM has been increasingly recognized as an inflammatory disease and monocytes play a key role in propagating inflammation under hyperglycemic conditions. We hypothesize that high glucose dysregulates microRNAs to promote monocyte inflammatory activity, which may contribute to the pathogenesis of DM. THP-1 monocytes were cultured in normal (5 mM) and high (25 mM) glucose conditions. RT-qPCR and Western blotting were performed to assay microRNAs and proteins, respectively. Monocytes were transfected with microRNA mimics using Lipofectamine RNAiMAX reagent. THP-1 monocyte growth was assessed using Calcein-AM dye and a Boyden chamber assay was applied to measure monocyte migration. The results showed that high glucose downregulated miR-139-5p associated with increased protein expression of CXCR4, an experimentally validated target of miR-139-5p. Correspondingly, treatment with high glucose resulted in a significant increase in THP-1 cell migration towards SDF-1, a cognate ligand for CXCR4. MiR-139-5p overexpression inhibited high glucose-induced CXCR4 expression, leading to reduced cell migration towards SDF-1. High glucose did not affect THP-1 monocyte growth. In conclusion, the miR-139-5p-CXCR4 axis may play a role in high glucose-induced inflammation by regulating monocyte migration.
Collapse
Affiliation(s)
- Weifang Li
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Gengchen Xu
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Gregory W Chai
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Alexander Ball
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Qiuwang Zhang
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada.
| | - Michael J B Kutryk
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
5
|
Santos AS, Santos-Bezerra DP, Ferreira LRP, Bando SY, Alves LI, Cunha-Neto E, da Silva MER. Relevance of Circulating microRNA, and their Association with Islet Cell Autoantibodies in Type 1 Diabetes Pathogenesis. Arch Med Res 2025; 56:103114. [PMID: 39489115 DOI: 10.1016/j.arcmed.2024.103114] [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: 05/30/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND AIMS/HYPOTHESIS The role of microRNAs (miRNAs) in the pathogenesis and progression of type 1 diabetes (T1D) has been described, but data remain scarce and conflicting. OBJECTIVES To evaluate the potential biological involvement of miRNA expression in the immune response and beta cell function in T1D. METHODS We screened 10 serum miRNAs from 142 subjects divided into three groups: healthy individuals (control group; n = 52) and patients at different stages of T1D progression, from the initial immunological manifestation, presenting islet cell autoantibodies (AbP group; n = 39), to partial and severe beta cell damage in T1D (recent T1D group; n = 51). RESULTS Three miRNAs (miR-200c-3p, miR-301a-3p, and miR-382-5p) were highly expressed in the AbP and/or recent T1D groups compared to the control group. Furthermore, in the AbP group, miR-301a-3p and miR-382-5p were positively correlated with insulin autoantibody levels and miR-382-5p was negatively correlated with C-peptide levels. In the recent T1D group, miR-200c-3p expression was positively correlated with IA-2A levels. Enrichment analysis of differentially expressed miRNAs showed their involvement in immune response, inflammatory pathways, proliferation/survival/apoptosis mechanisms, bacterial and viral infection, and insulin resistance. CONCLUSION Our data indicated that miR-200c-3p, miR-301a-3p, and miR-382-5p might be involved in T1D pathogenesis. Proliferative, metabolic, and immune responses were main pathways associated with serum miRNA target genes.
Collapse
Affiliation(s)
- Aritania S Santos
- Laboratorio de Carboidratos e Radioimunoensaios, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Ludmila Rodrigues Pinto Ferreira
- Department of Morphology, RNA Systems Biology Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Silvia Y Bando
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Laís Isidoro Alves
- Laboratorio de Carboidratos e Radioimunoensaios, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, Institute for Investigation in Immunology (iii) INCT, São Paulo, Brazil
| | - Maria Elizabeth Rossi da Silva
- Laboratorio de Carboidratos e Radioimunoensaios, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
6
|
Aljani B, Lindner A, Weigelt M, Zhao M, Sharma V, Bonifacio E, Jones P, Eugster A. Small RNA-Seq and real time rt-qPCR reveal islet miRNA released under stress conditions. Islets 2024; 16:2392343. [PMID: 39154325 PMCID: PMC11332650 DOI: 10.1080/19382014.2024.2392343] [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: 04/29/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024] Open
Abstract
Replacement of beta cells through transplantation is a potential therapeutic approach for individuals with pancreas removal or poorly controllable type 1 diabetes. However, stress and death of beta cells pose significant challenges. Circulating miRNA has emerged as potential biomarkers reflecting early beta cell stress and death, allowing for timely intervention. The aim of this study was to identify miRNAs as potential biomarkers for beta cell health. Literature review combined with small RNA sequencing was employed to select islet-enriched miRNA. The release of those miRNA was assessed by RT-qPCR in vivo, using a streptozotocin induced diabetes mouse model and in vitro, through mouse and human islets exposed to varying degrees of hypoxic and cytokine stressors. Utilizing the streptozotocin induced model, we identified 18 miRNAs out of 39 candidate islet-enriched miRNA to be released upon islet stress in vivo. In vitro analysis of culture supernatants from cytokine and/or hypoxia stressed islets identified the release of 45 miRNAs from mouse and 8 miRNAs from human islets. Investigation into the biological pathways targeted by the cytokine- and/or hypoxia-induced miRNA suggested the involvement of MAPK and PI3K-Akt signaling pathways in both mouse and human islets. We have identified miRNAs associated with beta cell health and stress. The findings allowed us to propose a panel of 47 islet-related human miRNA that is potentially valuable for application in clinical contexts of beta cell transplantation and presymptomatic early-stage type 1 diabetes.
Collapse
Affiliation(s)
- Bssam Aljani
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Annett Lindner
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Marc Weigelt
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Min Zhao
- German Center for Environmental Health, Institute of Diabetes Research, Helmholtz Munich, Munich, Germany
| | - Virag Sharma
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Ezio Bonifacio
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
- Faculty of Medicine, German Center for Diabetes Research (DZD), Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden, Dresden, Germany
| | - Peter Jones
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London, London, UK
| | - Anne Eugster
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| |
Collapse
|
7
|
Lécuyer E, Sauvageau M, Kothe U, Unrau PJ, Damha MJ, Perreault J, Abou Elela S, Bayfield MA, Claycomb JM, Scott MS. Canada's contributions to RNA research: past, present, and future perspectives. Biochem Cell Biol 2024; 102:472-491. [PMID: 39320985 DOI: 10.1139/bcb-2024-0176] [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: 09/27/2024] Open
Abstract
The field of RNA research has provided profound insights into the basic mechanisms modulating the function and adaption of biological systems. RNA has also been at the center stage in the development of transformative biotechnological and medical applications, perhaps most notably was the advent of mRNA vaccines that were critical in helping humanity through the Covid-19 pandemic. Unbeknownst to many, Canada boasts a diverse community of RNA scientists, spanning multiple disciplines and locations, whose cutting-edge research has established a rich track record of contributions across various aspects of RNA science over many decades. Through this position paper, we seek to highlight key contributions made by Canadian investigators to the RNA field, via both thematic and historical viewpoints. We also discuss initiatives underway to organize and enhance the impact of the Canadian RNA research community, particularly focusing on the creation of the not-for-profit organization RNA Canada ARN. Considering the strategic importance of RNA research in biology and medicine, and its considerable potential to help address major challenges facing humanity, sustained support of this sector will be critical to help Canadian scientists play key roles in the ongoing RNA revolution and the many benefits this could bring about to Canada.
Collapse
Affiliation(s)
- Eric Lécuyer
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, Canada
- Département de Biochimie et de Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
| | - Martin Sauvageau
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, Canada
- Département de Biochimie et de Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | - Ute Kothe
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada
| | - Peter J Unrau
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Masad J Damha
- Department of Chemistry, McGill University, Montréal, QC, Canada
| | - Jonathan Perreault
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, QC, Canada
| | - Sherif Abou Elela
- Département de Microbiologie et Infectiologie, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Julie M Claycomb
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Michelle S Scott
- Département de Biochimie et de Génomique Fonctionnelle, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
8
|
Li S, Zhou Y, Li K, Liu L, Fang M, Gao H. Inhibition of circDGKZ ameliorates myocardial ischemia/reperfusion injury by targeting miR-345-5p/TLR4. ESC Heart Fail 2024; 11:2730-2741. [PMID: 38725137 PMCID: PMC11424286 DOI: 10.1002/ehf2.14809] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/25/2024] [Accepted: 04/01/2024] [Indexed: 09/27/2024] Open
Abstract
AIMS This study aims to explore the molecular mechanism of circular RNAs' (circRNAs) potential involvement in myocardial ischaemia-reperfusion injury (MIRI). METHODS AND RESULTS Differently expressed genes in myocardial infarction (MI) were identified by screening the GEO database. Serum was collected from MI patients and healthy volunteers (n = 5 for each group). AC16 cells were cultured and exposed to hypoxia/reperfusion (H/R) treatment for the cell experiments. Then candidate genes were validated in human serum and the H/R model. Quantitative real-time PCR and western blot were used to detect expression of key molecules such as circDGKZ, miR-345-5p, and Toll-like receptor 4 (TLR4), as well as pyroptosis markers such as NOD-like receptor thermal protein domain-associated protein 3 (NLRP3), ASC, C-caspase1, interleukin (IL)-1β, and IL-18. CircDGKZ was positively correlated in human serum (P < 0.05) and in AC16 cells (P < 0.01). Knockdown of circDGKZ inhibited cardiomyocyte pyroptosis and the TLR4/nuclear factor kappa B (NF-κB) signalling pathway (all P < 0.05). A luciferase assay was used to detect the molecule interaction. MiR-345-5p was regulated by circDGKZ and regulated TLR4 in cardiomyocytes both through direct interaction (P < 0.01). The stability and distribution of circRNA or linear RNA were examined by subcellular localization and RNA decay assays. CircDGKZ was stably expressed in cardiomyocytes and mainly distributed in the cytoplasm (P < 0.01). Knockdown of circDGKZ also promoted the degradation of NLRP3 by inducing autophagy (P < 0.05). MIRI rat models were constructed (n = 5 for each group), and the cellular results were further confirmed in rat models (P < 0.05). CONCLUSIONS Knockdown of circDGKZ interrupted pyroptosis and induced autophagy of cardiomyocytes via regulating miR-345-5p/TLR4/NF-κB.
Collapse
Affiliation(s)
- Shiliang Li
- Division of Cardiothoracic and Vascular SurgeryTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - Yan Zhou
- Department of OtolaryngologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - KunSheng Li
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Lu Liu
- Department of PharmacyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - Ming Fang
- Health Management CenterUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - Hongfeng Gao
- Department of Emergency MedicineWuhan Wuchang Hospital, Wuchang Hospital Affiliated to Wuhan University of Science and TechnologyNo. 116, Yangyuan StreetWuhan430000Hubei ProvinceChina
| |
Collapse
|
9
|
Cabiati M, Federico G, Del Ry S. Importance of Studying Non-Coding RNA in Children and Adolescents with Type 1 Diabetes. Biomedicines 2024; 12:1988. [PMID: 39335501 PMCID: PMC11429055 DOI: 10.3390/biomedicines12091988] [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: 06/25/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Type 1 diabetes (T1D) mellitus is a chronic illness in children and teens, with rising global incidence rates. It stems from an autoimmune attack on pancreatic β cells, leading to insufficient insulin production. Genetic susceptibility and environmental triggers initiate this process. Early detection is possible by identifying multiple autoantibodies, which aids in predicting future T1D development. A new staging system highlights T1D's onset with islet autoimmunity rather than symptoms. Family members of T1D patients face a significantly increased risk of T1D. Italy recently passed a law mandating national T1D screening for pediatric populations. Measurements of β cell function continue to be essential in assessing efficacy, and different models have been proposed, but more appropriate biomarkers are mandatory for both progression studies before the onset of diabetes and during therapeutic monitoring. Biomarkers like microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) play key roles in T1D pathogenesis by regulating gene expression. Understanding their roles offers insights into T1D mechanisms and potential therapeutic targets. In this review, we summarized recent progress in the roles of some non-coding RNAs (ncRNAs) in the pathogenesis of T1D, with particular attention to miRNAs, lncRNAs, and circRNAs.
Collapse
Affiliation(s)
- Manuela Cabiati
- Laboratory of Biochemistry and Molecular Biology, Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Giovanni Federico
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Silvia Del Ry
- Laboratory of Biochemistry and Molecular Biology, Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| |
Collapse
|
10
|
Sebastiani G, Grieco GE, Bruttini M, Auddino S, Mori A, Toniolli M, Fignani D, Licata G, Aiello E, Nigi L, Formichi C, Fernandez-Tajes J, Pugliese A, Evans-Molina C, Overbergh L, Tree T, Peakman M, Mathieu C, Dotta F. A set of circulating microRNAs belonging to the 14q32 chromosome locus identifies two subgroups of individuals with recent-onset type 1 diabetes. Cell Rep Med 2024; 5:101591. [PMID: 38838677 PMCID: PMC11228666 DOI: 10.1016/j.xcrm.2024.101591] [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: 10/19/2023] [Revised: 02/02/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
Circulating microRNAs (miRNAs) are linked to the onset and progression of type 1 diabetes mellitus (T1DM), thus representing potential disease biomarkers. In this study, we employed a multiplatform sequencing approach to analyze circulating miRNAs in an extended cohort of prospectively evaluated recent-onset T1DM individuals from the INNODIA consortium. Our findings reveal that a set of miRNAs located within T1DM susceptibility chromosomal locus 14q32 distinguishes two subgroups of individuals. To validate our results, we conducted additional analyses on a second cohort of T1DM individuals, confirming the identification of these subgroups, which we have named cluster A and cluster B. Remarkably, cluster B T1DM individuals, who exhibit increased expression of a set of 14q32 miRNAs, show better glycemic control and display a different blood immunomics profile. Our findings suggest that this set of circulating miRNAs can identify two different T1DM subgroups with distinct blood immunomics at baseline and clinical outcomes during follow-up.
Collapse
Affiliation(s)
- Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Giuseppina Emanuela Grieco
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Marco Bruttini
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy; Tuscany Centre for Precision Medicine (CReMeP), Siena, Italy
| | - Stefano Auddino
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Alessia Mori
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy; Tuscany Centre for Precision Medicine (CReMeP), Siena, Italy
| | - Mattia Toniolli
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Daniela Fignani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Giada Licata
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Elena Aiello
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Caterina Formichi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | | | - Alberto Pugliese
- Diabetes Research Institute, Leonard Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Diabetes Immunology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases and the Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lut Overbergh
- Katholieke Universiteit Leuven/Universitaire Ziekenhuizen, Leuven, Belgium
| | - Timothy Tree
- Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Mark Peakman
- Immunology & Inflammation Research Therapeutic Area, Sanofi, Boston, MA, USA
| | - Chantal Mathieu
- Katholieke Universiteit Leuven/Universitaire Ziekenhuizen, Leuven, Belgium
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy; Tuscany Centre for Precision Medicine (CReMeP), Siena, Italy.
| |
Collapse
|
11
|
Daamouch S, Blüher M, Vázquez DC, Hackl M, Hofbauer LC, Rauner M. MiR-144-5p and miR-21-5p do not drive bone disease in a mouse model of type 1 diabetes mellitus. JBMR Plus 2024; 8:ziae036. [PMID: 38606150 PMCID: PMC11008730 DOI: 10.1093/jbmrpl/ziae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
The increased risk of fractures in patients with type 1 diabetes mellitus (T1DM) is nowadays well recognized. However, the exact mechanism of action of diabetic bone disease has not been fully elucidated. MicroRNAs (miRNAs) are gene regulators that operate post-transcriptionally and have been implicated in the development of various metabolic disorders including T1DM. Previous studies have implicated a role for miR-144-5p and miR-21-5p, which are involved in controlling oxidative stress by targeting Nrf2, in T1DM. To date, it is unclear whether miR-144-5p and miR-21-5p affect bone health in T1DM. Thus, this study aimed to investigate the influence of miR-144-5p and miR-21-5p knockdown in the development of bone disease in T1DM male mice. Therefore, T1DM was induced in 10-wk-old male mice using streptozotocin (STZ). One week later, after development of hyperglycemia, antagomir-144-5p and antagomir-21-5p or their non-targeting control were administered at 10 mg/kg BW once a week until the end of the experiment. At 14 wk of age, glucose levels, bone, and fat mass were analyzed. The results revealed that treating T1DM male mice with antagomir-144-5p and antagomir-21-5p did not protect against diabetes development or bone loss, despite the successful downregulation of the miRNAs and the normalization of Nrf2 mRNA levels in bone tissue. Histological and serological parameters of bone formation or resorption were not altered by the antagomir treatment. Finally, we measured the expression of miRNA-144-5p or miRNA-21-5p in the serum of 30 individuals with T1DM and compared them to non-diabetic controls, but did not find an altered expression of either miRNA. In conclusion, the knockdown of miR-144-5p and miR-21-5p does not affect STZ-induced diabetes development or loss of bone mass in male mice. However, it does normalize expression of the anti-oxidant factor Nrf2 in diabetic bone tissue.
Collapse
Affiliation(s)
- Souad Daamouch
- Department of Medicine III, Center for Healthy Aging, Technische Universität Dresden, Dresden, Saxony, 01307, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Saxony, 04109, Germany
| | | | | | - Lorenz C Hofbauer
- Department of Medicine III, Center for Healthy Aging, Technische Universität Dresden, Dresden, Saxony, 01307, Germany
| | - Martina Rauner
- Department of Medicine III, Center for Healthy Aging, Technische Universität Dresden, Dresden, Saxony, 01307, Germany
| |
Collapse
|
12
|
Gomez-Muñoz L, Dominguez-Bendala J, Pastori RL, Vives-Pi M. Immunometabolic biomarkers for partial remission in type 1 diabetes mellitus. Trends Endocrinol Metab 2024; 35:151-163. [PMID: 37949732 DOI: 10.1016/j.tem.2023.10.005] [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: 09/01/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023]
Abstract
Shortly after diagnosis of type 1 diabetes mellitus (T1DM) and initiation of insulin therapy, many patients experience a transient partial remission (PR) phase, also known as the honeymoon phase. This phase presents a potential therapeutic opportunity due to its association with immunoregulatory and β cell-protective mechanisms. However, the lack of biomarkers makes its characterization difficult. In this review, we cover the current literature addressing the discovery of new predictive and monitoring biomarkers that contribute to the understanding of the metabolic, epigenetic, and immunological mechanisms underlying PR. We further discuss how these peripheral biomarkers reflect attempts to arrest β cell autoimmunity and how these can be applied in clinical practice.
Collapse
Affiliation(s)
- Laia Gomez-Muñoz
- Immunology Section, Germans Trias i Pujol Research Institute, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ricardo L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Marta Vives-Pi
- Immunology Section, Germans Trias i Pujol Research Institute, Universitat Autònoma de Barcelona, 08916 Badalona, Spain; Ahead Therapeutics SL, 08193, Bellaterra, Barcelona, Spain.
| |
Collapse
|
13
|
Pang H, Fan W, Pi L, Shi X, Wang Z, Luo S, Xiao Y, Li X, Huang G, Xie Z, Zhou Z. Plasma-derived exosomal miRNA profiles associated with type 1 diabetes. Diabetes Metab Res Rev 2024; 40:e3774. [PMID: 38340050 DOI: 10.1002/dmrr.3774] [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: 05/24/2023] [Revised: 12/02/2023] [Accepted: 12/26/2023] [Indexed: 02/12/2024]
Abstract
AIMS Recently, exosomal miRNAs have been shown to play important roles in multiple diseases, including type 1 diabetes (T1D). To assess the biomarker potential of exosomal miRNAs for T1D, we measured the expression profiles of plasma-derived exosomal miRNAs in T1D and explored their potential functions by bioinformatic analysis. MATERIALS AND METHODS In the discovery phase, exosome samples were isolated from plasma by size exclusion chromatography from 10 T1D patients and 10 sex- (p = 0.36), age- (p = 0.97), and body mass index-matched (p = 0.47) healthy control subjects. Exosomal miRNA expression profiles were measured using the Illumina NovaSeq 6000 platform. With verification by quantitative real-time PCR (qRT-PCR), we used multiple bioinformatics approaches to explore the potential biological functions of the identified differentially expressed miRNAs. The diagnostic signature of exosomal miRNAs was evaluated by least absolute shrinkage and selection operator (LASSO) regression and evaluated based on the area under the receiver operating characteristic curve (AUC). RESULTS In total, 43 differentially expressed miRNAs, among which 34 were upregulated and 9 were downregulated, were identified in T1D. After correcting for multiple testing using false discovery rate, 11 identified exosomal miRNAs still showed statistical significance. Among the 5 selected miRNAs, 3 miRNAs (miR-103a-3p, miR-144-5p and miR-454-3p) were successfully validated by qRT-PCR. The biological analysis-enriched terms included protein autophosphorylation and the Hedgehog signalling pathway. The highest AUC of exosomal miRNA was 0.889 under the LASSO model. The expression levels of 5 selected exosomal miRNAs were correlated with multiple clinical characteristics such as fasting C-peptide and postprandial C-peptide. CONCLUSIONS Our results indicated that plasma-derived exosomal miRNAs could serve as promising diagnostic biomarkers of T1D.
Collapse
Affiliation(s)
- Haipeng Pang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wenqi Fan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Linhua Pi
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiajie Shi
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhen Wang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shuoming Luo
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yang Xiao
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xia Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Gan Huang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiguo Xie
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
14
|
Sassi G, Licata G, Ventriglia G, Wouters A, Lemaitre P, Seurinck R, Mori A, Grieco GE, Bissenova S, Ellis D, Caluwaerts S, Rottiers P, Vandamme N, Mathieu C, Dotta F, Gysemans C, Sebastiani G. A Plasma miR-193b-365 Signature Combined With Age and Glycemic Status Predicts Response to Lactococcus lactis-Based Antigen-Specific Immunotherapy in New-Onset Type 1 Diabetes. Diabetes 2023; 72:1470-1482. [PMID: 37494666 PMCID: PMC10545562 DOI: 10.2337/db22-0852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 07/10/2023] [Indexed: 07/28/2023]
Abstract
Immunomodulation combined with antigen therapy holds great promise to arrest autoimmune type 1 diabetes, but clinical translation is hampered by a lack of prognostic biomarkers. Low-dose anti-CD3 plus Lactococcus lactis bacteria secreting proinsulin and IL-10 reversed new-onset disease in nonobese diabetic (NOD) mice, yet some mice were resistant to the therapy. Using miRNA profiling, six miRNAs (i.e., miR-34a-5p, miR-125a-3p, miR-193b-3p, miR-328, miR-365-3p, and miR-671-3p) were identified as differentially expressed in plasma of responder versus nonresponder mice before study entry. After validation and stratification in an independent cohort, plasma miR-193b-3p and miR-365-3p, combined with age and glycemic status at study entry, had the best power to predict, with high sensitivity and specificity, poor response to the therapy. These miRNAs were highly abundant in pancreas-infiltrating neutrophils and basophils with a proinflammatory and activated phenotype. Here, a set of miRNAs and disease-associated parameters are presented as a predictive signature for the L. lactis-based immunotherapy outcome in new-onset type 1 diabetes, hence allowing targeted recruitment of trial participants and accelerated trial execution. ARTICLE HIGHLIGHTS Low-dose anti-CD3 combined with oral gavage of genetically modified Lactococcus lactis bacteria secreting human proinsulin and IL-10 holds great promise to arrest autoimmune type 1 diabetes, but the absence of biomarkers predicting therapeutic success hampers clinical translation. A set of cell-free circulation miRNAs together with age and glycemia at baseline predicts a poor response after L. lactis-based immunotherapy in nonobese mice with new-onset diabetes. Pancreas-infiltrating neutrophils and basophils are identified as potential cellular sources of discovered miRNAs. The prognostic signature could guide targeted recruitment of patients with newly diagnosed type 1 diabetes in clinical trials with the L. lactis-based immunotherapy.
Collapse
Affiliation(s)
- Gabriele Sassi
- Clinical and Experimental Endocrinology, Chrometa, KU Leuven, Leuven, Belgium
| | - Giada Licata
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario ONLUS, Toscana Life Science, Siena, Italy
| | - Giuliana Ventriglia
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario ONLUS, Toscana Life Science, Siena, Italy
| | - Amber Wouters
- Clinical and Experimental Endocrinology, Chrometa, KU Leuven, Leuven, Belgium
| | - Pierre Lemaitre
- Clinical and Experimental Endocrinology, Chrometa, KU Leuven, Leuven, Belgium
| | - Ruth Seurinck
- Data Mining and Modelling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Alessia Mori
- Tuscany Centre for Precision Medicine, Siena, Italy
| | - Giuseppina Emanuela Grieco
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario ONLUS, Toscana Life Science, Siena, Italy
| | - Samal Bissenova
- Clinical and Experimental Endocrinology, Chrometa, KU Leuven, Leuven, Belgium
| | - Darcy Ellis
- Clinical and Experimental Endocrinology, Chrometa, KU Leuven, Leuven, Belgium
| | | | | | - Niels Vandamme
- Data Mining and Modelling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
- VIB Single Cell Core, Leuven–Ghent, Ghent, Belgium
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, Chrometa, KU Leuven, Leuven, Belgium
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario ONLUS, Toscana Life Science, Siena, Italy
- Tuscany Centre for Precision Medicine, Siena, Italy
| | - Conny Gysemans
- Clinical and Experimental Endocrinology, Chrometa, KU Leuven, Leuven, Belgium
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario ONLUS, Toscana Life Science, Siena, Italy
| |
Collapse
|
15
|
Hara Y, Mizukami H, Yamazaki K, Yamada T, Igawa A, Takeuchi Y, Sasaki T, Kushibiki H, Murakami K, Kudoh K, Ishido K, Hakamada K. Dual epigenetic changes in diabetes mellitus-associated pancreatic ductal adenocarcinoma correlate with downregulation of E-cadherin and worsened prognosis. J Pathol Clin Res 2023; 9:354-366. [PMID: 37246239 PMCID: PMC10397378 DOI: 10.1002/cjp2.326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/02/2023] [Accepted: 05/04/2023] [Indexed: 05/30/2023]
Abstract
Diabetes mellitus (DM) is a risk factor for pancreatic ductal adenocarcinoma (PDAC) that promotes the promoter methylation of CDH1. It is still unclear whether DM can exert other epigenetic effects, such as altering microRNA (miR) expression, in PDAC. The expression of miR-100-5p is known to be changed in DM patients and can suppress the expression of E-cadherin. In this study, the correlation between DM status and dual epigenetic changes was evaluated in PDAC specimens from patients who underwent radical surgical resection. A total of 132 consecutive patients with PDAC were clinicopathologically evaluated. E-cadherin and nuclear β-catenin expression was measured using immunohistochemistry. DNA and miRs were extracted from the main tumor site on formalin-fixed paraffin-embedded tissue sections. TaqMan miR assays were applied to assess miR-100-5p expression. Bisulfite modification was conducted on the extracted DNA, which was then subjected to methylation-specific polymerase chain reaction. Immunohistochemistry revealed that decreased E-cadherin expression and increased nuclear β-catenin expression were significantly associated with DM and poor tumor cell differentiation. The presence of long-duration DM (≥3 years) was a significant factor contributing to CDH1 promoter methylation (p < 0.01), while miR-100-5p expression was proportionally correlated with the preoperative HbA1c level (R = 0.34, p < 0.01), but not the duration of DM. The subjects with high miR-100-5p expression and CDH1 promoter methylation showed the highest level of vessel invasion and prevalence of tumor size ≥30 mm. PDAC subjects with dual epigenetic changes showed poorer overall survival (OS) than those with a single epigenetic change. miR-100-5p expression ≥4.13 and CDH1 promoter methylation independently predicted poor OS and disease-free survival (DFS) in the multivariate analysis. OS and DFS worsened in DM subjects with both HbA1c ≥ 6.5% and DM duration ≥3 years. Thus, DM is associated with two modes of epigenetic change by independent mechanisms and worsens prognosis.
Collapse
Affiliation(s)
- Yutaro Hara
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| | - Hiroki Mizukami
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Keisuke Yamazaki
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| | - Takahiro Yamada
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| | - Akiko Igawa
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| | - Yuki Takeuchi
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Takanori Sasaki
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Hanae Kushibiki
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Kotaro Murakami
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Kazuhiro Kudoh
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Keinosuke Ishido
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| | - Kenichi Hakamada
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| |
Collapse
|
16
|
Jie R, Qian J, Tang Y, Li Y, Xu M, Zhao X, Chen M. Role of Increased miR-222-3p Expression in Peripheral Blood and Wound Marginal Tissues of Type 2 Diabetes Mellitus Patients with Diabetic Foot Ulcer. Diabetes Metab Syndr Obes 2023; 16:2419-2432. [PMID: 37602205 PMCID: PMC10439793 DOI: 10.2147/dmso.s410986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/28/2023] [Indexed: 08/22/2023] Open
Abstract
Purpose To study the correlations of miR-222-3p expression in the peripheral blood and wound marginal tissues of type 2 diabetes mellitus (T2DM) patients with the onset of diabetic foot ulcer (DFU), as well as explore the clinical value possessed by miR-222-3p in the diagnosis and treatment outcomes of DFU. Methods The study included 70 T2DM patients who did not suffer foot ulcers (T2DM group), 146 T2DM patients who suffered foot ulcers (DFU group), as well as 70 normal controls (NC group). Quantitative real-time PCR determined the MiR-222-3p relative expression. Clinical features and risk factors regarding DFU were assessed. Multiple stepwise logistic regression analysis assisted in confirming whether miR-222-3p expression could serve for independently predicting the risk factors for DFU. ROC curve analysis evaluated the diagnostic value exhibited by miR-222-3p level against DFU. Results T2DM group exhibited an obviously higher MiR-222-3p expression relative to NC group [1.98 (0.98, 3.62) vs 0.92 (0.61, 1.87)] (P < 0.01), but DFU group exhibited an obviously higher miR-222-3p expression relative to T2DM group [5.61 (1.98, 10.24) vs 1.98 (0.98, 3.62)] (P < 0.01). Besides, miR-222-3p expression presented a negative correlation with DFU healing rate (P < 0.05). According to Kaplan-Meier survival curve analysis, the group with high miR-222-3p expression showed higher unhealed DFU cumulative rate relative to the group with low expression (log-rank, P = 0.011, 0.001, respectively). Multivariate logistic regression analysis confirmed that high miR-222-3p expressions could independently predict DFU risk (OR=3.85, 95% CI 1.18~12.37, P = 0.008). According to the ROC curve analysis, the AUC of miR-222-3p specific to DFU diagnosis reached 0.803, with the best sensitivity of 95.93% and best specificity of 96.27%. Conclusion The increased expression of miR-222-3p in the peripheral blood of T2DM patients is closely related to the occurrence of DFU. MiR-222-3p is a biomarker with potential clinical value in diagnosing and evaluating the prognosis of DFU.
Collapse
Affiliation(s)
- Ruyan Jie
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Jing Qian
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Ying Tang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Yutong Li
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Murong Xu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Xiaotong Zhao
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Mingwei Chen
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| |
Collapse
|
17
|
Macvanin MT, Gluvic Z, Bajic V, Isenovic ER. Novel insights regarding the role of noncoding RNAs in diabetes. World J Diabetes 2023; 14:958-976. [PMID: 37547582 PMCID: PMC10401459 DOI: 10.4239/wjd.v14.i7.958] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 05/01/2023] [Accepted: 05/22/2023] [Indexed: 07/12/2023] Open
Abstract
Diabetes mellitus (DM) is a group of metabolic disorders defined by hyperglycemia induced by insulin resistance, inadequate insulin secretion, or excessive glucagon secretion. In 2021, the global prevalence of diabetes is anticipated to be 10.7% (537 million people). Noncoding RNAs (ncRNAs) appear to have an important role in the initiation and progression of DM, according to a growing body of research. The two major groups of ncRNAs implicated in diabetic disorders are miRNAs and long noncoding RNAs. miRNAs are single-stranded, short (17-25 nucleotides), ncRNAs that influence gene expression at the post-transcriptional level. Because DM has reached epidemic proportions worldwide, it appears that novel diagnostic and therapeutic strategies are required to identify and treat complications associated with these diseases efficiently. miRNAs are gaining attention as biomarkers for DM diagnosis and potential treatment due to their function in maintaining physiological homeostasis via gene expression regulation. In this review, we address the issue of the gradually expanding global prevalence of DM by presenting a complete and up-to-date synopsis of various regulatory miRNAs involved in these disorders. We hope this review will spark discussion about ncRNAs as prognostic biomarkers and therapeutic tools for DM. We examine and synthesize recent research that used novel, high-throughput technologies to uncover ncRNAs involved in DM, necessitating a systematic approach to examining and summarizing their roles and possible diagnostic and therapeutic uses.
Collapse
Affiliation(s)
- Mirjana T Macvanin
- Department of Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| | - Zoran Gluvic
- Department of Endocrinology and Diabetes, Clinic for Internal Medicine, Zemun Clinical Hospital, School of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Vladan Bajic
- Department of Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| | - Esma R Isenovic
- Department of Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| |
Collapse
|
18
|
Morales-Sánchez P, Lambert C, Ares-Blanco J, Suárez-Gutiérrez L, Villa-Fernández E, Garcia AV, García-Villarino M, Tejedor JR, Fraga MF, Torre EM, Pujante P, Delgado E. Circulating miRNA expression in long-standing type 1 diabetes mellitus. Sci Rep 2023; 13:8611. [PMID: 37244952 DOI: 10.1038/s41598-023-35836-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023] Open
Abstract
Type 1 diabetes is a chronic autoimmune disease which results in inefficient regulation of glucose homeostasis and can lead to different vascular comorbidities through life. In this study we aimed to analyse the circulating miRNA expression profile of patients with type 1 diabetes, and with no other associated pathology. For this, fasting plasma was obtained from 85 subjects. Next generation sequencing analysis was firstly performed to identify miRNAs that were differentially expressed between groups (20 patients vs. 10 controls). hsa-miR-1-3p, hsa-miR-200b-3p, hsa-miR-9-5p, and hsa-miR-1200 expression was also measured by Taqman RT-PCR to validate the observed changes (34 patients vs. 21 controls). Finally, through a bioinformatic approach, the main pathways affected by the target genes of these miRNAs were studied. Among the studied miRNAs, hsa-miR-1-3p expression was found significantly increased in patients with type 1 diabetes compared to controls, and positively correlated with glycated haemoglobin levels. Additionally, by using a bioinformatic approach, we could observe that changes in hsa-miR-1-3p directly affect genes involved in vascular development and cardiovascular pathologies. Our results suggest that, circulating hsa-miR-1-3p in plasma, together with glycaemic control, could be used as prognostic biomarkers in type 1 diabetes, helping to prevent the development of vascular complications in these patients.
Collapse
Affiliation(s)
- Paula Morales-Sánchez
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Lambert
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- University of Barcelona, Barcelona, Spain.
| | - Jessica Ares-Blanco
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
| | - Lorena Suárez-Gutiérrez
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - Elsa Villa-Fernández
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Ana Victoria Garcia
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Miguel García-Villarino
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Juan Ramón Tejedor
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Oviedo, Asturias, Spain
- Institute of Oncology of Asturias (IUOPA), Oviedo, Asturias, Spain
- Department of Organisms and Systems Biology (B.O.S), University of Oviedo, Oviedo, Asturias, Spain
| | - Mario F Fraga
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Oviedo, Asturias, Spain
- Institute of Oncology of Asturias (IUOPA), Oviedo, Asturias, Spain
- Department of Organisms and Systems Biology (B.O.S), University of Oviedo, Oviedo, Asturias, Spain
| | - Edelmiro Menéndez Torre
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
| | - Pedro Pujante
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain.
| | - Elías Delgado
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain.
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain.
| |
Collapse
|
19
|
Li H, Hu X, Li J, Jiang W, Wang L, Tan X. Identification of key regulatory genes and their working mechanisms in type 1 diabetes. BMC Med Genomics 2023; 16:8. [PMID: 36650594 PMCID: PMC9843847 DOI: 10.1186/s12920-023-01432-y] [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: 07/04/2022] [Accepted: 01/04/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease characterized by the destruction of beta cells in pancreatic islets. Identification of the key genes involved in T1D progression and their mechanisms of action may contribute to a better understanding of T1D. METHODS The microarray profile of T1D-related gene expression was searched using the Gene Expression Omnibus (GEO) database. Then, the expression data of two messenger RNAs (mRNAs) were integrated for Weighted Gene Co-Expression Network Analysis (WGCNA) to generate candidate genes related to T1D. In parallel, T1D microRNA (miRNA) data were analyzed to screen for possible regulatory miRNAs and their target genes. An miRNA-mRNA regulatory network was then established to predict the key regulatory genes and their mechanisms. RESULTS A total of 24 modules (i.e., clusters/communities) were selected using WGCNA analysis, in which three modules were significantly associated with T1D. Further correlation analysis of the gene module revealed 926 differentially expressed genes (DEGs), of which 327 genes were correlated with T1D. Analysis of the miRNA microarray showed that 13 miRNAs had significant expression differences in T1D. An miRNA-mRNA network was established based on the prediction of miRNA target genes and the combined analysis of mRNA, in which the target genes of two miRNAs were found in T1D correlated genes. CONCLUSION An miRNA-mRNA network for T1D was established, based on which 2 miRNAs and 12 mRNAs were screened, suggesting that they may play key regulatory roles in the initiation and development of T1D.
Collapse
Affiliation(s)
- Hui Li
- grid.508008.50000 0004 4910 8370Pediatric Department, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410000 Hunan People’s Republic of China
| | - Xiao Hu
- grid.508008.50000 0004 4910 8370Pediatric Department, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410000 Hunan People’s Republic of China
| | - Jieqiong Li
- grid.508008.50000 0004 4910 8370Pediatric Department, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410000 Hunan People’s Republic of China
| | - Wen Jiang
- grid.508008.50000 0004 4910 8370Pediatric Department, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410000 Hunan People’s Republic of China
| | - Li Wang
- grid.508008.50000 0004 4910 8370Pediatric Department, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410000 Hunan People’s Republic of China
| | - Xin Tan
- grid.508008.50000 0004 4910 8370Pediatric Department, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410000 Hunan People’s Republic of China
| |
Collapse
|
20
|
Phowira J, Ahmed FW, Bakhashab S, Weaver JU. Upregulated miR-18a-5p in Colony Forming Unit-Hill’s in Subclinical Cardiovascular Disease and Metformin Therapy; MERIT Study. Biomedicines 2022; 10:biomedicines10092136. [PMID: 36140236 PMCID: PMC9496122 DOI: 10.3390/biomedicines10092136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Colony forming unit-Hill’s (CFU-Hill’s) colonies are hematopoietic-derived cells that participate in neovasculogenesis and serve as a biomarker for vascular health. In animals, overexpression of miR-18a-5p was shown to be pro-atherogenic. We had shown that well-controlled type 1 diabetes mellitus (T1DM) is characterized by an inflammatory state, endothelial dysfunction, and reduced number of CFU-Hill’s, a model of subclinical cardiovascular disease (CVD). MERIT study explored the role of miR-18a-5p expression in CFU-Hill’s colonies in T1DM, and the cardioprotective effect of metformin in subclinical CVD. In T1DM, miR-18a-5p was significantly upregulated whereas metformin reduced it to HC levels. MiR-18a-5p was inversely correlated with CFU-Hill’s colonies, CD34+, CD34+CD133+ cells, and positively with IL-10, C-reactive protein, vascular endothelial growth factor-D (VEGF-D), and thrombomodulin. The receiver operating characteristic curve demonstrated, miR-18a-5p as a biomarker of T1DM, and upregulated miR-18a-5p defining subclinical CVD at HbA1c of 44.5 mmol/mol (pre-diabetes). Ingenuity pathway analysis documented miR-18a-5p inhibiting mRNA expression of insulin-like growth factor-1, estrogen receptor-1, hypoxia-inducible factor-1α cellular communication network factor-2, and protein inhibitor of activated STAT 3, whilst metformin upregulated these mRNAs via transforming growth factor beta-1 and VEGF. We confirmed the pro-atherogenic effect of miR-18a-5p in subclinical CVD and identified several target genes for future CVD therapies.
Collapse
Affiliation(s)
- Jason Phowira
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Fahad W. Ahmed
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle upon Tyne NE9 6SH, UK
- Department of Medical Oncology, King Faisal Specialist Hospital and Research Centre, Madinah 42522, Saudi Arabia
| | - Sherin Bakhashab
- Biochemistry Department, King Abdulaziz University, P.O. Box 80218, Jeddah 21589, Saudi Arabia
| | - Jolanta U. Weaver
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle upon Tyne NE9 6SH, UK
- Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Correspondence: ; Tel.: +44-191-445-2181
| |
Collapse
|
21
|
Ohlstrom DJ, Sul C, Vohwinkel CU, Hernandez-Lagunas L, Karimpour-Fard A, Mourani PM, Carpenter TC, Nozik ES, Sucharov CC. Plasma microRNA and metabolic changes associated with pediatric acute respiratory distress syndrome: a prospective cohort study. Sci Rep 2022; 12:14560. [PMID: 36028738 PMCID: PMC9418138 DOI: 10.1038/s41598-022-15476-0] [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] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 06/24/2022] [Indexed: 11/09/2022] Open
Abstract
Acute respiratory distress syndrome is a heterogeneous pathophysiological process responsible for significant morbidity and mortality in pediatric intensive care patients. Diagnosis is defined by clinical characteristics that identify the syndrome after development. Subphenotyping patients at risk of progression to ARDS could provide the opportunity for therapeutic intervention. microRNAs, non-coding RNAs stable in circulation, are a promising biomarker candidate. We conducted a single-center prospective cohort study to evaluate random forest classification of microarray-quantified circulating microRNAs in critically ill pediatric patients. We additionally selected a sub-cohort for parallel metabolomics profiling as a pilot study for concurrent use of miRNAs and metabolites as circulating biomarkers. In 35 patients (n = 21 acute respiratory distress, n = 14 control) 15 microRNAs were differentially expressed. Unsupervised random forest classification accurately grouped ARDS and control patients with an area under the curve of 0.762, which was improved to 0.839 when subset to only patients with bacterial infection. Nine metabolites were differentially abundant between acute respiratory distress and control patients (n = 4, both groups) and abundance was highly correlated with miRNA expression. Random forest classification of microRNAs differentiated critically ill pediatric patients who developed acute respiratory distress relative to those who do not. The differential expression of microRNAs and metabolites provides a strong foundation for further work to validate their use as a prognostic biomarker.
Collapse
Affiliation(s)
- Denis J Ohlstrom
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Christina Sul
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Division of Pediatric Critical Care, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Christine U Vohwinkel
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Division of Pediatric Critical Care, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Laura Hernandez-Lagunas
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Division of Pediatric Critical Care, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Anis Karimpour-Fard
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Peter M Mourani
- Division of Pediatric Critical Care, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Section of Pediatric Critical Care, Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Todd C Carpenter
- Division of Pediatric Critical Care, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Eva S Nozik
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Division of Pediatric Critical Care, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Carmen C Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado, Anschutz Medical Campus, 12700 E 19th Ave B139, Aurora, CO, 80045, USA.
| |
Collapse
|
22
|
Progression of Type 1 Diabetes: Circulating MicroRNA Expression Profiles Changes from Preclinical to Overt Disease. J Immunol Res 2022; 2022:2734490. [PMID: 35903753 PMCID: PMC9325579 DOI: 10.1155/2022/2734490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/01/2022] [Indexed: 11/17/2022] Open
Abstract
Objectives To evaluate the potential biological involvement of miRNA expression in the immune response and beta cell function in T1D. Methods We screened 377 serum miRNAs of 110 subjects divided into four groups: healthy individuals (control group) and patients at different stages of T1D progression, from the initial immunological manifestation presenting islet autoantibodies (AbP group) until partial and strong beta cell damage in the recent (recent T1D group) and long-term T1D, with 2 to 5 years of disease (T1D 2-5y group). Results The results revealed 69 differentially expressed miRNAs (DEMs) in relation to controls. Several miRNAs were correlated with islet autoantibodies (IA2A, GADA, and Znt8A), age, and C-peptide levels, mainly from AbP, and recent T1D groups pointing these miRNAs as relevant to T1D pathogenesis and progression. Several miRNAs were related to metabolic derangements, inflammatory pathways, and several other autoimmune diseases. Pathway analysis of putative DEM targets revealed an enrichment in pathways related to metabolic syndrome, inflammatory response, apoptosis and insulin signaling pathways, metabolic derangements, and decreased immunomodulation. One of the miRNAs' gene targets was DYRK2 (dual-specificity tyrosine-phosphorylation-regulated kinase 2), which is an autoantigen targeted by an antibody in T1D. ROC curve analysis showed hsa-miR-16 and hsa-miR-200a-3p with AUCs greater than for glucose levels, with discriminating power for T1D prediction greater than glucose levels. Conclusions/Interpretation. Our data suggests a potential influence of DEMs on disease progression from the initial autoimmune lesion up to severe beta cell dysfunction and the role of miRNAs hsa-miR-16 and hsa-miR-200a-3p as biomarkers of T1D progression.
Collapse
|
23
|
Paolini A, Baldassarre A, Bruno SP, Felli C, Muzi C, Ahmadi Badi S, Siadat SD, Sarshar M, Masotti A. Improving the Diagnostic Potential of Extracellular miRNAs Coupled to Multiomics Data by Exploiting the Power of Artificial Intelligence. Front Microbiol 2022; 13:888414. [PMID: 35756065 PMCID: PMC9218639 DOI: 10.3389/fmicb.2022.888414] [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: 03/02/2022] [Accepted: 05/11/2022] [Indexed: 12/15/2022] Open
Abstract
In recent years, the clinical use of extracellular miRNAs as potential biomarkers of disease has increasingly emerged as a new and powerful tool. Serum, urine, saliva and stool contain miRNAs that can exert regulatory effects not only in surrounding epithelial cells but can also modulate bacterial gene expression, thus acting as a “master regulator” of many biological processes. We think that in order to have a holistic picture of the health status of an individual, we have to consider comprehensively many “omics” data, such as miRNAs profiling form different parts of the body and their interactions with cells and bacteria. Moreover, Artificial Intelligence (AI) and Machine Learning (ML) algorithms coupled to other multiomics data (i.e., big data) could help researchers to classify better the patient’s molecular characteristics and drive clinicians to identify personalized therapeutic strategies. Here, we highlight how the integration of “multiomic” data (i.e., miRNAs profiling and microbiota signature) with other omics (i.e., metabolomics, exposomics) analyzed by AI algorithms could improve the diagnostic and prognostic potential of specific biomarkers of disease.
Collapse
Affiliation(s)
- Alessandro Paolini
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | | | - Stefania Paola Bruno
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy.,Department of Science, University Roma Tre, Rome, Italy
| | - Cristina Felli
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Chantal Muzi
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Sara Ahmadi Badi
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.,Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.,Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Meysam Sarshar
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Andrea Masotti
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| |
Collapse
|
24
|
Garavelli S, Prattichizzo F, Ceriello A, Galgani M, de Candia P. Type 1 Diabetes and Associated Cardiovascular Damage: Contribution of Extracellular Vesicles in Tissue Crosstalk. Antioxid Redox Signal 2022; 36:631-651. [PMID: 34407376 DOI: 10.1089/ars.2021.0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Type 1 diabetes (T1D) is characterized by the autoimmune destruction of the insulin secreting β-cells, with consequent aberrant blood glucose levels. Hyperglycemia is the common denominator for most of the chronic diabetic vascular complications, which represent the main cause of life reduction in T1D patients. For this disease, three interlaced medical needs remain: understanding the underlying mechanisms involved in pancreatic β-cell loss; identifying biomarkers able to predict T1D progression and its related complications; recognizing novel therapeutic targets. Recent Advances: Extracellular vesicles (EVs), released by most cell types, were discovered to contain a plethora of different molecules (including microRNAs) with regulatory properties, which are emerging as mediators of cell-to-cell communication at the paracrine and endocrine level. Recent knowledge suggests that EVs may act as pathogenic factors, and be developed into disease biomarkers and therapeutic targets in the context of several human diseases. Critical Issues: EVs have been recently shown to sustain a dysregulated cellular crosstalk able to exacerbate the autoimmune response in the pancreatic islets of T1D; moreover, EVs were shown to be able to monitor and/or predict the progression of T1D and the insurgence of vasculopathies. Future Directions: More mechanistic studies are needed to investigate whether the dysregulation of EVs in T1D patients is solely reflecting the progression of diabetes and related complications, or EVs also directly participate in the disease process, thus pointing to a potential use of EVs as therapeutic targets/tools in T1D. Antioxid. Redox Signal. 36, 631-651.
Collapse
Affiliation(s)
- Silvia Garavelli
- Institute for Endocrinology and Experimental Oncology "G. Salvatore," Consiglio Nazionale delle Ricerche (C.N.R.), Naples, Italy
| | | | | | - Mario Galgani
- Institute for Endocrinology and Experimental Oncology "G. Salvatore," Consiglio Nazionale delle Ricerche (C.N.R.), Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II," Italy
| | | |
Collapse
|
25
|
Barseem NF, Mahasab MM, Zaed IF, Said AEA, El Gayed EMA. Genetic Indices Relationship to Hyperglycemia-associated Biomarkers: Consistency with miRNA Expression in Egyptian Children with T1DM. J Clin Res Pediatr Endocrinol 2022; 14:76-86. [PMID: 34927407 PMCID: PMC8900082 DOI: 10.4274/jcrpe.galenos.2021.2021.0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE Micro RNAs (miRNAs) are gaining acceptance as novel biomarkers for the autoimmune disorders. However, miRNA profiles have not been investigated in individuals at risk of or diagnosed with type 1 diabetes mellitus (T1DM). To study the expression pattern of miRNAs in plasma obtained from patients with T1DM and compare with matched healthy controls. METHODS Equal numbers of patients with T1DM (90) and healthy-matched control children (90) were assessed for the expression profile of plasma miRNAs including miRNA-101-5p, miRNA-146-5p, miRNA-21-5p, miRNA-375, miRNA-126, and Let7a-5p using reverse transcriptase polymerase chain reaction methodology and quantitative real-time testing. RESULTS Analysis showed that miRNA-101, miRNA-21 and miRNA-375 were highly expressed, whereas, miRNA-146-5p, miRNA-126, and miRNA-Let7a-5p showed significantly low levels of expression in T1DM patients compared to controls (p<0.05). In addition, miRNA-101 and miRNA-146 correlated with age at diagnosis of T1DM and disease duration, respectively. Furthermore, multivariate analysis showed that miRNA-126 and Let7a-5p had a significant negative correlation with mean hemoglobin A1c (HbA1c) values. CONCLUSION Dysregulation of the six miRNAs analyzed suggested a possible role as biomarkers in T1DM. miRNA-101 was correlated with age at diagnosis while miRNA-146 correlated with disease duration. Two further miRNAs correlated with the existing biomarker, HbA1c.
Collapse
Affiliation(s)
- Naglaa Fathy Barseem
- Menoufia University Faculty of Medicine, Department of Pediatric, Unit of Genetic and Endocrinology, Shebein Elkom, Egypt,* Address for Correspondence: Menoufia University Faculty of Medicine, Department of Pediatric, Unit of Genetic and Endocrinology, Shebein Elkom, Egypt Phone: +00201000314896 E-mail:
| | - Marwa Mohamed Mahasab
- Menoufia University Faculty of Medicine, Department of Family Medicine, Shebein Elkom, Egypt
| | - Ibrahem Fathy Zaed
- Menoufia University Faculty of Sciences, Department of Chemistry, Shebein Elkom, Egypt
| | - Aya Eldesoky A. Said
- Menoufia University Faculty of Sciences, Department of Chemistry, Shebein Elkom, Egypt
| | - Eman Masoud Abd El Gayed
- Menoufia University Faculty of Medicine, Department of Medical Biochemistry and Molecular Biology, Shebein Elkom, Egypt
| |
Collapse
|
26
|
Mukherjee S, Shelar B, Krishna S. Versatile role of miR-24/24-1*/24-2* expression in cancer and other human diseases. Am J Transl Res 2022; 14:20-54. [PMID: 35173828 PMCID: PMC8829624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/08/2021] [Indexed: 06/14/2023]
Abstract
MiRNAs (miRs) have been proven to be well-validated therapeutic targets. Emerging evidence has demonstrated that intricate, intrinsic and paradoxical functions of miRs are context-dependent because of their multiple upstream regulators, broad spectrum of downstream molecular targets and distinct expression in various tissues, organs and disease states. Targeted therapy has become an emerging field of research. One key for the development of successful miR-based/targeted therapy is to acquire integrated knowledge of its regulatory network and its association with disease phenotypes to identify critical nodes of the underlying pathogenesis. Herein, we systematically summarized the comprehensive role of miR-24-3p (miR-24), along with its passenger strands miR-24-1-5p* (miR-24-1) and miR-24-2-5p* (miR-24-2), emphasizing their microenvironment, intracellular targets, and associated gene networks and regulatory phenotypes in 18 different cancer types and 13 types of other disorders. MiR-24 targets and regulates numerous genes in various cancer types and enhances the expression of several oncogenes (e.g., cMyc, BCL2 and HIF1), which are challenging in terms of druggability. In contrast, several tumor suppressor proteins (p21 and p53) have been reported to be downregulated by miR-24. MiR-24 also regulates the cell cycle and is associated with numerous cancer hallmarks such as apoptosis, proliferation, metastasis, invasion, angiogenesis, autophagy, drug resistance and other diseases pathogenesis. Overall, miR-24 plays an emerging role in the diagnosis, prognosis and pathobiology of various diseases. MiR-24 is a potential target for targeted therapy in the era of precision medicine, which expands the landscape of targetable macromolecules, including undruggable proteins.
Collapse
Affiliation(s)
| | | | - Sudhir Krishna
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR)Bellary Road, Bangalore 560065, Karnataka, India
| |
Collapse
|
27
|
Micro-RNA Implications in Type-1 Diabetes Mellitus: A Review of Literature. Int J Mol Sci 2021; 22:ijms222212165. [PMID: 34830046 PMCID: PMC8621893 DOI: 10.3390/ijms222212165] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/24/2021] [Accepted: 11/04/2021] [Indexed: 02/07/2023] Open
Abstract
Type-1 diabetes mellitus (T1DM) is one of the most well-defined and complex metabolic disorders, characterized by hyperglycemia, with a constantly increasing incidence in children and adolescents. While current knowledge regarding the molecules related to the pathogenesis and diagnosis of T1DM is vast, the discovery of new molecules, such as micro ribonucleic acids (micro-RNAs, miRNAs), as well as their interactions with T1DM, has spurred novel prospects in the diagnosis of the disease. This review aims at summarizing current knowledge regarding miRNAs' biosynthesis and action pathways and their role as gene expression regulators in T1DM. MiRNAs follow a complex biosynthesis pathway, including cleaving and transport from nucleus to cytoplasm. After assembly of their final form, they inhibit translation or cause messenger RNA (mRNA) degradation, resulting in the obstruction of protein synthesis. Many studies have reported miRNA involvement in T1DM pathogenesis, mainly through interference with pancreatic b-cell function, insulin production and secretion. They are also found to contribute to β-cell destruction, as they aid in the production of autoreactive agents. Due to their elevated accumulation in various biological specimens, as well as their involvement in T1DM pathogenesis, their role as biomarkers in early preclinical T1DM diagnosis is widely hypothesized, with future studies concerning their diagnostic value deemed a necessity.
Collapse
|
28
|
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
|
29
|
Frørup C, Mirza AH, Yarani R, Nielsen LB, Mathiesen ER, Damm P, Svare J, Engelbrekt C, Størling J, Johannesen J, Mortensen HB, Pociot F, Kaur S. Plasma Exosome-Enriched Extracellular Vesicles From Lactating Mothers With Type 1 Diabetes Contain Aberrant Levels of miRNAs During the Postpartum Period. Front Immunol 2021; 12:744509. [PMID: 34691048 PMCID: PMC8531745 DOI: 10.3389/fimmu.2021.744509] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes is an immune-driven disease, where the insulin-producing beta cells from the pancreatic islets of Langerhans becomes target of immune-mediated destruction. Several studies have highlighted the implication of circulating and exosomal microRNAs (miRNAs) in type 1 diabetes, underlining its biomarker value and novel therapeutic potential. Recently, we discovered that exosome-enriched extracellular vesicles carry altered levels of both known and novel miRNAs in breast milk from lactating mothers with type 1 diabetes. In this study, we aimed to characterize exosomal miRNAs in the circulation of lactating mothers with and without type 1 diabetes, hypothesizing that differences in type 1 diabetes risk in offspring from these groups are reflected in the circulating miRNA profile. We performed small RNA sequencing on exosome-enriched extracellular vesicles extracted from plasma of 52 lactating mothers around 5 weeks postpartum (26 with type 1 diabetes and 26 age-matched controls), and found a total of 2,289 miRNAs in vesicles from type 1 diabetes and control libraries. Of these, 176 were differentially expressed in plasma from mothers with type 1 diabetes (167 upregulated; 9 downregulated, using a cut-off of abs(log2FC) >1 and FDR adjusted p-value <0.05). Extracellular vesicles were verified by nanoparticle tracking analysis, transmission electron microscopy and immunoblotting. Five candidate miRNAs were selected based on their involvement in diabetes and immune modulation/beta-cell functions: hsa-miR-127-3p, hsa-miR-146a-5p, hsa-miR-26a-5p, hsa-miR-24-3p and hsa-miR-30d-5p. Real-time qPCR validation confirmed that hsa-miR-146a-5p, hsa-miR-26a-5p, hsa-miR-24-3p, and hsa-miR-30d-5p were significantly upregulated in lactating mothers with type 1 diabetes as compared to lactating healthy mothers. To determine possible target genes and affected pathways of the 5 miRNA candidates, computational network-based analyses were carried out with TargetScan, mirTarBase, QIAGEN Ingenuity Pathway Analysis and PantherDB database. The candidates showed significant association with inflammatory response and cytokine and chemokine mediated signaling pathways. With this study, we detect aberrant levels of miRNAs within plasma extracellular vesicles from lactating mothers with type 1 diabetes during the postpartum period, including miRNAs with associations to disease pathogenesis and inflammatory responses.
Collapse
Affiliation(s)
- Caroline Frørup
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aashiq H Mirza
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Pharmacology, Weill Cornell Medical, New York, NY, United States
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Lotte B Nielsen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Elisabeth R Mathiesen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Peter Damm
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Center for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen, Denmark.,Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
| | - Jens Svare
- Department of Obstetrics, Herlev and Gentofte Hospital, Herlev, Denmark
| | | | - Joachim Størling
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Johannesen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Henrik B Mortensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Simranjeet Kaur
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| |
Collapse
|
30
|
Kahn SE, Chen YC, Esser N, Taylor AJ, van Raalte DH, Zraika S, Verchere CB. The β Cell in Diabetes: Integrating Biomarkers With Functional Measures. Endocr Rev 2021; 42:528-583. [PMID: 34180979 PMCID: PMC9115372 DOI: 10.1210/endrev/bnab021] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 02/08/2023]
Abstract
The pathogenesis of hyperglycemia observed in most forms of diabetes is intimately tied to the islet β cell. Impairments in propeptide processing and secretory function, along with the loss of these vital cells, is demonstrable not only in those in whom the diagnosis is established but typically also in individuals who are at increased risk of developing the disease. Biomarkers are used to inform on the state of a biological process, pathological condition, or response to an intervention and are increasingly being used for predicting, diagnosing, and prognosticating disease. They are also proving to be of use in the different forms of diabetes in both research and clinical settings. This review focuses on the β cell, addressing the potential utility of genetic markers, circulating molecules, immune cell phenotyping, and imaging approaches as biomarkers of cellular function and loss of this critical cell. Further, we consider how these biomarkers complement the more long-established, dynamic, and often complex measurements of β-cell secretory function that themselves could be considered biomarkers.
Collapse
Affiliation(s)
- Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Yi-Chun Chen
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Nathalie Esser
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Austin J Taylor
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Daniël H van Raalte
- Department of Internal Medicine, Amsterdam University Medical Center (UMC), Vrije Universiteit (VU) University Medical Center, 1007 MB Amsterdam, The Netherlands.,Department of Experimental Vascular Medicine, Amsterdam University Medical Center (UMC), Academic Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Sakeneh Zraika
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - C Bruce Verchere
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| |
Collapse
|
31
|
Belli R, Ferraro E, Molfino A, Carletti R, Tambaro F, Costelli P, Muscaritoli M. Liquid Biopsy for Cancer Cachexia: Focus on Muscle-Derived microRNAs. Int J Mol Sci 2021; 22:ijms22169007. [PMID: 34445710 PMCID: PMC8396502 DOI: 10.3390/ijms22169007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer cachexia displays a complex nature in which systemic inflammation, impaired energy metabolism, loss of muscle and adipose tissues result in unintentional body weight loss. Cachectic patients have a poor prognosis and the presence of cachexia reduces the tolerability of chemo/radio-therapy treatments and it is frequently the primary cause of death in advanced cancer patients. Early detection of this condition could make treatments more effective. However, early diagnostic biomarkers of cachexia are currently lacking. In recent years, although solid biopsy still remains the "gold standard" for diagnosis of cancer, liquid biopsy is gaining increasing interest as a source of easily accessible potential biomarkers. Moreover, the growing interest in circulating microRNAs (miRNAs), has made these molecules attractive for the diagnosis of several diseases, including cancer. Some muscle-derived circulating miRNA might play a pivotal role in the onset/progression of cancer cachexia. This topic is of great interest since circulating miRNAs might be easily detectable by means of liquid biopsies and might allow an early diagnosis of this syndrome. We here summarize the current knowledge on circulating muscular miRNAs involved in muscle atrophy, since they might represent easily accessible and promising biomarkers of cachexia.
Collapse
Affiliation(s)
- Roberta Belli
- Department of Translational and Precision Medicine, Sapienza University, 00185 Rome, Italy; (A.M.); (R.C.); (F.T.)
- Correspondence: (R.B.); (M.M.); Tel./Fax: +390-649-972-020 (M.M.)
| | - Elisabetta Ferraro
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56126 Pisa, Italy;
| | - Alessio Molfino
- Department of Translational and Precision Medicine, Sapienza University, 00185 Rome, Italy; (A.M.); (R.C.); (F.T.)
| | - Raffaella Carletti
- Department of Translational and Precision Medicine, Sapienza University, 00185 Rome, Italy; (A.M.); (R.C.); (F.T.)
| | - Federica Tambaro
- Department of Translational and Precision Medicine, Sapienza University, 00185 Rome, Italy; (A.M.); (R.C.); (F.T.)
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Torino, 10124 Torino, Italy;
| | - Maurizio Muscaritoli
- Department of Translational and Precision Medicine, Sapienza University, 00185 Rome, Italy; (A.M.); (R.C.); (F.T.)
- Correspondence: (R.B.); (M.M.); Tel./Fax: +390-649-972-020 (M.M.)
| |
Collapse
|
32
|
Margaritis K, Margioula-Siarkou G, Margioula-Siarkou C, Petousis S, Kotanidou EP, Christoforidis A, Pavlou E, Galli-Tsinopoulou A. Circulating serum and plasma levels of micro-RNA in type-1 diabetes in children and adolescents: A systematic review and meta-analysis. Eur J Clin Invest 2021; 51:e13510. [PMID: 33565089 DOI: 10.1111/eci.13510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/26/2021] [Accepted: 02/05/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is a complex metabolic disorder characterized by hyperglycaemia, with constantly increasing incidence in paediatric population. The discovery of new molecules, such as microRNAs, and their possible interactions with T1DM create novel aspects in the diagnosis of the disease. METHODS This systematic review and meta-analysis adhered to PRISMA guidelines. MEDLINE, SCOPUS, Cochrane CENTRAL and Clinicaltrials.gov. were searched up to 20 April 2020. Inclusion criteria for individual studies were quantification of microRNAs in serum/plasma samples and study groups consisting of children and adolescents with T1DM and healthy controls. Primary outcome of the study was the qualitative expression of microRNAs between the two groups. Statistical analysis was performed with Comprehensive Meta-Analysis Software v3.0. Methodological quality of included studies was assessed using Newcastle-Ottawa scale. RESULTS A total of 484 studies were retrieved from the initial search of the databases. These were subsequently limited to seven included studies. Seven microRNAs demonstrated contrasting expression between the two groups, with two of them showing significant overexpression in T1DM group (miR-181:95% CI: 0.429 to 1.341 P < .001, miR-210:95% CI: 0.381 to 0.852, P < .001) and one micro-RNA being significantly overexpressed in control group (miR-375:95% CI: 0.293 to 1.459, P = .003). CONCLUSION A total of three micro-RNA molecules appeared to have a significantly different expression in T1DM patients, serving as a possible diagnostic panel of biomarkers. These findings may contribute as reference for future research to further support the use of microRNAs as a novel diagnostic tool in T1DM.
Collapse
Affiliation(s)
- Kosmas Margaritis
- 2nd Department of Paediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, AHEPA University General Hospital, Thessaloniki, Greece
| | - Georgia Margioula-Siarkou
- 2nd Department of Obstetrics and Gynecology, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, Thessaloniki, Greece
| | - Chrysoula Margioula-Siarkou
- 2nd Department of Obstetrics and Gynecology, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, Thessaloniki, Greece
| | - Stamatios Petousis
- 2nd Department of Obstetrics and Gynecology, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, Thessaloniki, Greece
| | - Eleni P Kotanidou
- 2nd Department of Paediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, AHEPA University General Hospital, Thessaloniki, Greece
| | - Athanasios Christoforidis
- 1st Department of Paediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, Thessaloniki, Greece
| | - Evangelos Pavlou
- 2nd Department of Paediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, AHEPA University General Hospital, Thessaloniki, Greece
| | - Assimina Galli-Tsinopoulou
- 2nd Department of Paediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, AHEPA University General Hospital, Thessaloniki, Greece
| |
Collapse
|
33
|
MicroRNAs-1299, -126-3p and -30e-3p as Potential Diagnostic Biomarkers for Prediabetes. Diagnostics (Basel) 2021; 11:diagnostics11060949. [PMID: 34073154 PMCID: PMC8226728 DOI: 10.3390/diagnostics11060949] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 12/13/2022] Open
Abstract
This cross-sectional study investigated the association of miR-1299, -126-3p and -30e-3p with and their diagnostic capability for dysglycaemia in 1273 (men, n = 345) South Africans, aged >20 years. Glycaemic status was assessed by oral glucose tolerance test (OGTT). Whole blood microRNA (miRNA) expressions were assessed using TaqMan-based reverse transcription quantitative-PCR (RT-qPCR). Receiver operating characteristic (ROC) curves assessed the ability of each miRNA to discriminate dysglycaemia, while multivariable logistic regression analyses linked expression with dysglycaemia. In all, 207 (16.2%) and 94 (7.4%) participants had prediabetes and type 2 diabetes mellitus (T2DM), respectively. All three miRNAs were significantly highly expressed in individuals with prediabetes compared to normotolerant patients, p < 0.001. miR-30e-3p and miR-126-3p were also significantly more expressed in T2DM versus normotolerant patients, p < 0.001. In multivariable logistic regressions, the three miRNAs were consistently and continuously associated with prediabetes, while only miR-126-3p was associated with T2DM. The ROC analysis indicated all three miRNAs had a significant overall predictive ability to diagnose prediabetes, diabetes and the combination of both (dysglycaemia), with the area under the receiver operating characteristic curve (AUC) being significantly higher for miR-126-3p in prediabetes. For prediabetes diagnosis, miR-126-3p (AUC = 0.760) outperformed HbA1c (AUC = 0.695), p = 0.042. These results suggest that miR-1299, -126-3p and -30e-3p are associated with prediabetes, and measuring miR-126-3p could potentially contribute to diabetes risk screening strategies.
Collapse
|
34
|
Vasu S, Yang JM, Hodges J, Abu-El-Haija MA, Adams DB, Balamurugan AN, Beilman GJ, Chinnakotla S, Conwell DL, Freeman ML, Gardner TB, Hatipoglu B, Kirchner V, Lara LF, Morgan KA, Nathan JD, Posselt A, Pruett TL, Schwarzenberg SJ, Singh VK, Wijkstrom M, Witkowski P, Naziruddin B, Bellin MD. Circulating miRNA in Patients Undergoing Total Pancreatectomy and Islet Autotransplantation. Cell Transplant 2021; 30:963689721999330. [PMID: 33902338 PMCID: PMC8718159 DOI: 10.1177/0963689721999330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Circulating microRNAs (miRNAs) can be biomarkers for diagnosis and progression of several pathophysiological conditions. In a cohort undergoing total pancreatectomy with islet autotransplantation (TPIAT) from the multicenter Prospective Observational Study of TPIAT (POST), we investigated associations between a panel of circulating miRNAs (hsa-miR-375, hsa-miR-29b-3p, hsa-miR-148a-3p, hsa-miR-216a-5p, hsa-miR-320d, hsa-miR-200c, hsa-miR-125b, hsa-miR-7-5p, hsa-miR-221-3p, hsa-miR-122-5p) and patient, disease and islet-isolation characteristics. Plasma samples (n = 139) were collected before TPIAT and miRNA levels were measured by RTPCR. Disease duration, prior surgery, and pre-surgical diabetes were not associated with circulating miRNAs. Levels of hsa-miR-29b-3p (P = 0.03), hsa-miR-148a-3p (P = 0.04) and hsa-miR-221-3p (P = 0.01) were lower in those with genetic risk factors. Levels of hsa-miR-148a-3p (P = 0.04) and hsa-miR-7-5p (P = 0.04) were elevated in toxic/metabolic disease. Participants with exocrine insufficiency had lower hsa-miR-29b-3p, hsa-miR-148a-3p, hsa-miR-320d, hsa-miR-221-3p (P < 0.01) and hsa-miR-375, hsa-miR-200c-3p, and hsa-miR-125b-5p (P < 0.05). Four miRNAs were associated with fasting C-peptide before TPIAT (hsa-miR-29b-3p, r = 0.18; hsa-miR-148a-3p, r = 0.21; hsa-miR-320d, r = 0.19; and hsa-miR-221-3p, r = 0.21; all P < 0.05), while hsa-miR-29b-3p was inversely associated with post-isolation islet equivalents/kg and islet number/kg (r = −0.20, P = 0.02). Also, hsa-miR-200c (r = 0.18, P = 0.03) and hsa-miR-221-3p (r = 0.19, P = 0.03) were associated with islet graft tissue volume. Further investigation is needed to determine the predictive potential of these miRNAs for assessing islet autotransplant outcomes.
Collapse
Affiliation(s)
- Srividya Vasu
- Islet Cell Laboratory, Baylor University Medical Center, Dallas TX, USA
| | - Jiemin M Yang
- School of Public Health (Biostatistics), University of Minnesota, Minneapolis, MN, USA
| | - James Hodges
- School of Public Health (Biostatistics), University of Minnesota, Minneapolis, MN, USA
| | | | - David B Adams
- Medical University of South Carolina, Charleston, SC, USA
| | - Appakalai N Balamurugan
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,University of Louisville, Louisville, KY, USA
| | - Greg J Beilman
- University of Minnesota Medical School, Minneapolis, MN, USA
| | | | - Darwin L Conwell
- The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | | | | | | | - Luis F Lara
- The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Jaimie D Nathan
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew Posselt
- University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | | | | - Bashoo Naziruddin
- Islet Cell Laboratory, Baylor University Medical Center, Dallas TX, USA
| | - Melena D Bellin
- University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
35
|
Lassandro G, Ciaccia L, Amoruso A, Palladino V, Palmieri VV, Giordano P. Focus on MicroRNAs as Biomarker in Pediatric Diseases. Curr Pharm Des 2021; 27:826-832. [PMID: 33087027 DOI: 10.2174/1381612826666201021125512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/25/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND MiRNAs are a class of small non-coding RNAs that are involved in the post-transcriptional regulation of gene expression. MiRNAs are considered a class of epigenetic biomarkers. These biomarkers can investigate disease at different stages: diagnosis, therapy or clinical follow-up. OBJECTIVE The aim of this paper is to highlight the innovative use of miRNAs in several childhood diseases. METHODS We conducted a literature review to search the usage of miRNAs in pediatric clinical routine or experimental trials. RESULTS We found a possible key role of miRNAs in different pediatric illnesses (metabolic alterations, coagulation defects, cancer). CONCLUSION The modest literature production denotes that further investigation is needed to assess and validate the promising role of miRNAs as non-invasive biomarkers in pediatric disorders.
Collapse
Affiliation(s)
- Giuseppe Lassandro
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Loredana Ciaccia
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Anna Amoruso
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Valentina Palladino
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Viviana V Palmieri
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Paola Giordano
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
36
|
Gradisteanu Pircalabioru G, Corcionivoschi N, Gundogdu O, Chifiriuc MC, Marutescu LG, Ispas B, Savu O. Dysbiosis in the Development of Type I Diabetes and Associated Complications: From Mechanisms to Targeted Gut Microbes Manipulation Therapies. Int J Mol Sci 2021; 22:2763. [PMID: 33803255 PMCID: PMC7967220 DOI: 10.3390/ijms22052763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Globally, we are facing a worrying increase in type 1 diabetes mellitus (T1DM) incidence, with onset at younger age shedding light on the need to better understand the mechanisms of disease and step-up prevention. Given its implication in immune system development and regulation of metabolism, there is no surprise that the gut microbiota is a possible culprit behind T1DM pathogenesis. Additionally, microbiota manipulation by probiotics, prebiotics, dietary factors and microbiota transplantation can all modulate early host-microbiota interactions by enabling beneficial microbes with protective potential for individuals with T1DM or at high risk of developing T1DM. In this review, we discuss the challenges and perspectives of translating microbiome data into clinical practice. Nevertheless, this progress will only be possible if we focus our interest on developing numerous longitudinal, multicenter, interventional and double-blind randomized clinical trials to confirm their efficacy and safety of these therapeutic approaches.
Collapse
Affiliation(s)
| | - Nicolae Corcionivoschi
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast BT9 5PX, UK;
| | - Ozan Gundogdu
- Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK;
| | - Mariana-Carmen Chifiriuc
- Research Institute of University of Bucharest (ICUB), 300645 Bucharest, Romania; (G.G.P.); (L.G.M.); (B.I.)
- Academy of Romanian Scientists, 050045 Bucharest, Romania
| | | | - Bogdan Ispas
- Research Institute of University of Bucharest (ICUB), 300645 Bucharest, Romania; (G.G.P.); (L.G.M.); (B.I.)
| | - Octavian Savu
- “N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 2nd District, 020042 Bucharest, Romania;
- Department of Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 5th District, 050474 Bucharest, Romania
| |
Collapse
|
37
|
Alcazar O, Hernandez LF, Nakayasu ES, Nicora CD, Ansong C, Muehlbauer MJ, Bain JR, Myer CJ, Bhattacharya SK, Buchwald P, Abdulreda MH. Parallel Multi-Omics in High-Risk Subjects for the Identification of Integrated Biomarker Signatures of Type 1 Diabetes. Biomolecules 2021; 11:383. [PMID: 33806609 PMCID: PMC7999903 DOI: 10.3390/biom11030383] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Biomarkers are crucial for detecting early type-1 diabetes (T1D) and preventing significant β-cell loss before the onset of clinical symptoms. Here, we present proof-of-concept studies to demonstrate the potential for identifying integrated biomarker signature(s) of T1D using parallel multi-omics. METHODS Blood from human subjects at high risk for T1D (and healthy controls; n = 4 + 4) was subjected to parallel unlabeled proteomics, metabolomics, lipidomics, and transcriptomics. The integrated dataset was analyzed using Ingenuity Pathway Analysis (IPA) software for disturbances in the at-risk subjects compared to controls. RESULTS The final quadra-omics dataset contained 2292 proteins, 328 miRNAs, 75 metabolites, and 41 lipids that were detected in all samples without exception. Disease/function enrichment analyses consistently indicated increased activation, proliferation, and migration of CD4 T-lymphocytes and macrophages. Integrated molecular network predictions highlighted central involvement and activation of NF-κB, TGF-β, VEGF, arachidonic acid, and arginase, and inhibition of miRNA Let-7a-5p. IPA-predicted candidate biomarkers were used to construct a putative integrated signature containing several miRNAs and metabolite/lipid features in the at-risk subjects. CONCLUSIONS Preliminary parallel quadra-omics provided a comprehensive picture of disturbances in high-risk T1D subjects and highlighted the potential for identifying associated integrated biomarker signatures. With further development and validation in larger cohorts, parallel multi-omics could ultimately facilitate the classification of T1D progressors from non-progressors.
Collapse
Affiliation(s)
- Oscar Alcazar
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
| | - Luis F. Hernandez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
| | - Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (E.S.N.); (C.D.N.); (C.A.)
| | - Carrie D. Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (E.S.N.); (C.D.N.); (C.A.)
| | - Charles Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (E.S.N.); (C.D.N.); (C.A.)
| | - Michael J. Muehlbauer
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; (M.J.M.); (J.R.B.)
| | - James R. Bain
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; (M.J.M.); (J.R.B.)
| | - Ciara J. Myer
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.J.M.); (S.K.B.)
- Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sanjoy K. Bhattacharya
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.J.M.); (S.K.B.)
- Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Peter Buchwald
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Midhat H. Abdulreda
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.J.M.); (S.K.B.)
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
38
|
Jakubik D, Fitas A, Eyileten C, Jarosz-Popek J, Nowak A, Czajka P, Wicik Z, Sourij H, Siller-Matula JM, De Rosa S, Postula M. MicroRNAs and long non-coding RNAs in the pathophysiological processes of diabetic cardiomyopathy: emerging biomarkers and potential therapeutics. Cardiovasc Diabetol 2021; 20:55. [PMID: 33639953 PMCID: PMC7916283 DOI: 10.1186/s12933-021-01245-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/13/2021] [Indexed: 02/08/2023] Open
Abstract
The epidemic of diabetes mellitus (DM) necessitates the development of novel therapeutic and preventative strategies to attenuate complications of this debilitating disease. Diabetic cardiomyopathy (DCM) is a frequent disorder affecting individuals diagnosed with DM characterized by left ventricular hypertrophy, diastolic and systolic dysfunction and myocardial fibrosis in the absence of other heart diseases. Progression of DCM is associated with impaired cardiac insulin metabolic signaling, increased oxidative stress, impaired mitochondrial and cardiomyocyte calcium metabolism, and inflammation. Various non-coding RNAs, such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), as well as their target genes are implicated in the complex pathophysiology of DCM. It has been demonstrated that miRNAs and lncRNAs play an important role in maintaining homeostasis through regulation of multiple genes, thus they attract substantial scientific interest as biomarkers for diagnosis, prognosis and as a potential therapeutic strategy in DM complications. This article will review the different miRNAs and lncRNA studied in the context of DM, including type 1 and type 2 diabetes and the contribution of pathophysiological mechanisms including inflammatory response, oxidative stress, apoptosis, hypertrophy and fibrosis to the development of DCM .
Collapse
Affiliation(s)
- Daniel Jakubik
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland
| | - Alex Fitas
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland
| | - Joanna Jarosz-Popek
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland.,Doctoral School, Medical University of Warsaw, 02-091, Warsaw, Poland
| | - Anna Nowak
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland
| | - Pamela Czajka
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland
| | - Zofia Wicik
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland.,Centro de Matemática, Computação e Cognição, Universidade Federal Do ABC, São Paulo, Brazil
| | - Harald Sourij
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Jolanta M Siller-Matula
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland.,Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Salvatore De Rosa
- Division of Cardiology, Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy.,Cardiovascular Research Center, "Magna Graecia" University, Catanzaro, Italy
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland.
| |
Collapse
|
39
|
Li YY, Zhang S, Wang H, Zhang SX, Xu T, Chen SW, Zhang Y, Chen Y. Identification of Crucial Genes and Pathways Associated with Atherosclerotic Plaque in Diabetic Patients. Pharmgenomics Pers Med 2021; 14:211-220. [PMID: 33568933 PMCID: PMC7869704 DOI: 10.2147/pgpm.s281705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/26/2020] [Indexed: 01/20/2023] Open
Abstract
Background Patients with diabetes have more calcification in atherosclerotic plaque and a higher occurrence of secondary cardiovascular events than patients without diabetes. The objective of this study was to identify crucial genes involved in the development of diabetic atherosclerotic plaque using a bioinformatics approach. Methods Microarray dataset GSE118481 was downloaded from the Gene Expression Omnibus (GEO) database; the dataset included 6 patients with diabetic atherosclerotic plaque (DBT) and 6 nondiabetic patients with atherosclerotic plaque (Ctrl). Differentially expressed genes (DEG) between the DBT and Ctrl groups were identified and then subjected to functional enrichment analysis. Based on the enriched pathways of DEGs, diabetic atherosclerotic plaque-related pathways were screened using the comparative toxicogenomics database (CTD). We then constructed a protein–protein interaction (PPI) network and transcription factor (TF)–miRNA–mRNA network. Results A total of 243 DEGs were obtained in the DBT group compared with the Ctrl group, including 85 up-regulated and 158 down-regulated DEGs. Functional enrichment analysis showed that up-regulated DEGs were mainly enriched in isoprenoid metabolic process, DNA-binding TF activity, and response to virus. Additionally, DEGs participating in the toll-like receptor signaling pathway were closely related to diabetes, carotid stenosis, and insulin resistance. The TF–miRNA–mRNA network showed that toll-like receptor 4 (TLR4), BCL2-like 11 (BCL2L11), and glutamate-cysteine ligase catalytic subunit (GCLC) were hub genes. Furthermore, TLR4 was regulated by TF signal transducer and activator of transcription 6 (STAT6); BCL2L11 was targeted by hsa-miR-24-3p; and GCLC was regulated by nuclear factor, erythroid 2 like 2 (NFE2L2). Conclusion Identification of hub genes and pathways increased our understanding of the molecular mechanisms underlying the atherosclerotic plaque in patients with or without diabetes. These crucial genes (TLR4, BC2L11, and GCLC) might function as molecular biomarkers for diabetic atherosclerotic plaque.
Collapse
Affiliation(s)
- Yuan-Yuan Li
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Sheng Zhang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Hua Wang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Shun-Xiao Zhang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Ting Xu
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Shu-Wen Chen
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Yan Zhang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Yue Chen
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| |
Collapse
|
40
|
Kwak YH, Kwak DK, Moon HS, Kim NY, Yee JS, Yoo JH. Significant Changes in Serum MicroRNAs after High Tibial Osteotomy in Medial Compartmental Knee Osteoarthritis: Potential Prognostic Biomarkers. Diagnostics (Basel) 2021; 11:258. [PMID: 33562261 PMCID: PMC7914593 DOI: 10.3390/diagnostics11020258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/21/2022] Open
Abstract
High tibial osteotomy (HTO) is an effective alternative for medial compartmental knee osteoarthritis (OA). Circulating microRNAs (miRNAs) are known to serve as OA-related biomarkers. The present study investigated the differential expression of serum miRNAs before and after HTO to identify potential miRNAs as prognostic biomarkers. miRNA-polymerase chain reaction (PCR) arrays were used to screen for miRNAs in the serum at preoperative and 6-month postoperative time points from six patients, and the differentially expressed miRNAs identified in the profiling stage were validated using real-time PCR at post-operative months 6 and 18 in 27 other HTO-treated patients. Among 84 miRNAs involved in the inflammatory process, three (miR-19b-3p, miR-29c-3p, and miR-424-5p) showed differential expression patterns in the profiling stage (p = 0.011, 0.015, and 0.021, respectively). Levels of these three and four other miRNAs (miR-140-3p, miR-454-3p, miR-let-7e-5p, and miR-885-5p) known to be related to OA progression were evaluated in the serum of 27 patients. Only four miRNAs (miR-19b-3p, miR-140-3p, miR-454-3p, and miR-let-7e-5p) were significantly upregulated at postoperative month 6 (p = 0.003, 0.005, 0.004, and 0.004, respectively), and only miR-140-3p was significantly upregulated up to 18 months after operation (p = 0.003). Together, this study reveals the significantly upregulated serum miRNAs after HTO as potential prognostic biomarkers; however, further studies are warranted to elucidate their clinical implications.
Collapse
Affiliation(s)
- Yoon Hae Kwak
- Division of Orthopaedic Surgery, Severance Children’s Hospital, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Dae-Kyung Kwak
- Department of Orthopaedic Surgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Korea; (D.-K.K.); (H.-S.M.); (J.-S.Y.)
| | - Hyun-Soo Moon
- Department of Orthopaedic Surgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Korea; (D.-K.K.); (H.-S.M.); (J.-S.Y.)
| | - Nan Young Kim
- Hallym Institute of Translational Genomics & Bioinformatics, Hallym University Medical Center, Anyang 14068, Korea;
| | - Jae-Sung Yee
- Department of Orthopaedic Surgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Korea; (D.-K.K.); (H.-S.M.); (J.-S.Y.)
| | - Je-Hyun Yoo
- Department of Orthopaedic Surgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Korea; (D.-K.K.); (H.-S.M.); (J.-S.Y.)
| |
Collapse
|
41
|
Snowhite I, Pastori R, Sosenko J, Messinger Cayetano S, Pugliese A. Baseline Assessment of Circulating MicroRNAs Near Diagnosis of Type 1 Diabetes Predicts Future Stimulated Insulin Secretion. Diabetes 2021; 70:638-651. [PMID: 33277338 PMCID: PMC7881864 DOI: 10.2337/db20-0817] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022]
Abstract
Type 1 diabetes is an autoimmune disease resulting in severely impaired insulin secretion. We investigated whether circulating microRNAs (miRNAs) are associated with residual insulin secretion at diagnosis and predict the severity of its future decline. We studied 53 newly diagnosed subjects enrolled in placebo groups of TrialNet clinical trials. We measured serum levels of 2,083 miRNAs, using RNA sequencing technology, in fasting samples from the baseline visit (<100 days from diagnosis), during which residual insulin secretion was measured with a mixed meal tolerance test (MMTT). Area under the curve (AUC) C-peptide and peak C-peptide were stratified by quartiles of expression of 31 miRNAs. After adjustment for baseline C-peptide, age, BMI, and sex, baseline levels of miR-3187-3p, miR-4302, and the miRNA combination of miR-3187-3p/miR-103a-3p predicted differences in MMTT C-peptide AUC/peak levels at the 12-month visit; the combination miR-3187-3p/miR-4723-5p predicted proportions of subjects above/below the 200 pmol/L clinical trial eligibility threshold at the 12-month visit. Thus, miRNA assessment at baseline identifies associations with C-peptide and stratifies subjects for future severity of C-peptide loss after 1 year. We suggest that miRNAs may be useful in predicting future C-peptide decline for improved subject stratification in clinical trials.
Collapse
Affiliation(s)
- Isaac Snowhite
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Ricardo Pastori
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
- Division of Endocrinology and Metabolism, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Jay Sosenko
- Division of Endocrinology and Metabolism, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Shari Messinger Cayetano
- Department of Public Health Sciences, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Alberto Pugliese
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
- Division of Endocrinology and Metabolism, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
- Department of Microbiology and Immunology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| |
Collapse
|
42
|
Paul S, Ruiz-Manriquez LM, Ledesma-Pacheco SJ, Benavides-Aguilar JA, Torres-Copado A, Morales-Rodríguez JI, De Donato M, Srivastava A. Roles of microRNAs in chronic pediatric diseases and their use as potential biomarkers: A review. Arch Biochem Biophys 2021; 699:108763. [PMID: 33460581 DOI: 10.1016/j.abb.2021.108763] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/22/2020] [Accepted: 01/10/2021] [Indexed: 02/09/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding highly conserved RNA molecules that can act as master regulators of gene expression in a sequence-specific manner either by translation repression or mRNA degradation, influencing a wide range of biologic processes that are essential for the maintenance of cellular homeostasis. Chronic pediatric diseases are the leading cause of death worldwide among children and the recent evidence indicates that aberrant miRNA expression significantly contributes to the development of chronic pediatric diseases. This review focuses on the role of miRNAs in five major chronic pediatric diseases including bronchial asthma, congenital heart diseases, cystic fibrosis, type 1 diabetes mellitus, and epilepsy, and their potential use as novel biomarkers for the diagnosis and prognosis of these disorders.
Collapse
Affiliation(s)
- Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico.
| | - Luis M Ruiz-Manriquez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - S Janin Ledesma-Pacheco
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - Javier A Benavides-Aguilar
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - Andrea Torres-Copado
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - Jonathan I Morales-Rodríguez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - Marcos De Donato
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - Aashish Srivastava
- Section of Bioinformatics, Clinical Laboratory, Haukeland University Hospital, Bergen, 5021, Norway; Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
| |
Collapse
|
43
|
Potential therapeutic approaches of microRNAs for COVID-19: Challenges and opportunities. J Oral Biol Craniofac Res 2020; 11:132-137. [PMID: 33398242 PMCID: PMC7772998 DOI: 10.1016/j.jobcr.2020.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/23/2020] [Indexed: 02/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) emerges as current outbreak cause by Novel Severe Acute Respiratory Syndrome Corona Virus-2 (SARS-CoV-2). This infection affects respiratory system and provides uncontrolled systemic inflammatory response as cytokine storm. The main concern about SARS-CoV-2 pandemic is high viral pathogenicity with no specific drugs. MicroRNAs (miRs) as small non-coding RNAs (21–25 nt) regulate gene expression. The SARS-CoV-2 encoded-miRs affect human genes that involved in transcription, translation, apoptosis, immune response and inflammation. Also, they alter self-gene regulation and hijacked host miRs that provide protective environment to maintain its latency. On the other hand, Host miRs play critical role in viral gene expression to restrict infection. Over expression/inhibition of miRs might result in cell cycle irregularity, impaired immune response or cancer. In this manner, exact role of each miR should be specified. Mimic encoded-miRs like antagomirs showed successful result in phases of clinical trial prevent from negative effects of viral encoded-miRs. Products of mimic miRs are inexpensive corresponds to synthesis of primer; they are short and nanoscale in size. Although SARS-CoV-2 genome is undergoing evaluation, detection of exact molecular pathogenesis open up opportunities to for vaccine development. Salivaomics can evaluate SARS-CoV-2 genome, transcriptome, proteome and biomarkers like miRs in oral related and cancer disease. In this review, we studied the challenge and opportunities of miRs in therapeutic approach for SARS-CoV-2 infection, then overviewed the role of miRs in saliva droplet during SARS-CoV-2 infection and related cancer.
Collapse
|
44
|
Garavelli S, Bruzzaniti S, Tagliabue E, Di Silvestre D, Prattichizzo F, Mozzillo E, Fattorusso V, La Sala L, Ceriello A, Puca AA, Mauri P, Strollo R, Marigliano M, Maffeis C, Petrelli A, Bosi E, Franzese A, Galgani M, Matarese G, de Candia P. Plasma circulating miR-23~27~24 clusters correlate with the immunometabolic derangement and predict C-peptide loss in children with type 1 diabetes. Diabetologia 2020; 63:2699-2712. [PMID: 32728892 DOI: 10.1007/s00125-020-05237-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS We aimed to analyse the association between plasma circulating microRNAs (miRNAs) and the immunometabolic profile in children with type 1 diabetes and to identify a composite signature of miRNAs/immunometabolic factors able to predict type 1 diabetes progression. METHODS Plasma samples were obtained from children at diagnosis of type 1 diabetes (n = 88) and at 12 (n = 32) and 24 (n = 30) months after disease onset and from healthy control children with similar sex and age distribution (n = 47). We quantified 60 robustly expressed plasma circulating miRNAs by quantitative RT-PCR and nine plasma immunometabolic factors with a recognised role at the interface of metabolic and immune alterations in type 1 diabetes. Based on fasting C-peptide loss over time, children with type 1 diabetes were stratified into the following groups: those who had lost >90% of C-peptide compared with diagnosis level; those who had lost <10% of C-peptide; those showing an intermediate C-peptide loss. To evaluate the modulation of plasma circulating miRNAs during the course of type 1 diabetes, logistic regression models were implemented and the correlation between miRNAs and immunometabolic factors was also assessed. Results were then validated in an independent cohort of children with recent-onset type 1 diabetes (n = 18). The prognostic value of the identified plasma signature was tested by a neural network-based model. RESULTS Plasma circulating miR-23~27~24 clusters (miR-23a-3p, miR-23b-3p, miR-24-3p, miR-27a-3p and miR-27b-3p) were upmodulated upon type 1 diabetes progression, showed positive correlation with osteoprotegerin (OPG) and were negatively correlated with soluble CD40 ligand, resistin, myeloperoxidase and soluble TNF receptor in children with type 1 diabetes but not in healthy children. The combination of plasma circulating miR-23a-3p, miR-23b-3p, miR-24-3p, miR-27b-3p and OPG, quantified at disease onset, showed a significant capability to predict the decline in insulin secretion 12 months after disease diagnosis in two independent cohorts of children with type 1 diabetes. CONCLUSIONS/INTERPRETATIONS We have pinpointed a novel miR-23a-3p/miR-23b-3p/miR-24-3p/miR-27b-3p/OPG plasma signature that may be developed into a novel blood-based method to better stratify patients with type 1 diabetes and predict C-peptide loss.
Collapse
Affiliation(s)
- Silvia Garavelli
- IRCCS MultiMedica, via G. Fantoli 16/15, 20138, Milan, Italy
- Institute for Endocrinology and Experimental Oncology 'G. Salvatore', C.N.R, via Pansini 5, 80131, Naples, Italy
| | - Sara Bruzzaniti
- Institute for Endocrinology and Experimental Oncology 'G. Salvatore', C.N.R, via Pansini 5, 80131, Naples, Italy
- Department of Biology, University of Naples 'Federico II', Naples, Italy
| | - Elena Tagliabue
- IRCCS MultiMedica, via G. Fantoli 16/15, 20138, Milan, Italy
| | | | | | - Enza Mozzillo
- Centre of Paediatric Diabetology, Department of Translational Medical Sciences, University of Naples 'Federico II', Naples, Italy
| | - Valentina Fattorusso
- Centre of Paediatric Diabetology, Department of Translational Medical Sciences, University of Naples 'Federico II', Naples, Italy
| | - Lucia La Sala
- IRCCS MultiMedica, via G. Fantoli 16/15, 20138, Milan, Italy
| | | | - Annibale A Puca
- IRCCS MultiMedica, via G. Fantoli 16/15, 20138, Milan, Italy
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Pierluigi Mauri
- Institute of Biomedical Technologies, C. N. R, Segrate, Milan, Italy
| | - Rocky Strollo
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico, Rome, Italy
| | - Marco Marigliano
- Paediatric Diabetes and Metabolic Disorders Unit, University of Verona, Verona, Italy
| | - Claudio Maffeis
- Paediatric Diabetes and Metabolic Disorders Unit, University of Verona, Verona, Italy
| | - Alessandra Petrelli
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Emanuele Bosi
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Adriana Franzese
- Centre of Paediatric Diabetology, Department of Translational Medical Sciences, University of Naples 'Federico II', Naples, Italy
| | - Mario Galgani
- Institute for Endocrinology and Experimental Oncology 'G. Salvatore', C.N.R, via Pansini 5, 80131, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II', via Pansini 5, 80131, Naples, Italy.
| | - Giuseppe Matarese
- Institute for Endocrinology and Experimental Oncology 'G. Salvatore', C.N.R, via Pansini 5, 80131, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II', via Pansini 5, 80131, Naples, Italy.
| | - Paola de Candia
- IRCCS MultiMedica, via G. Fantoli 16/15, 20138, Milan, Italy.
| |
Collapse
|
45
|
Reading between the (Genetic) Lines: How Epigenetics is Unlocking Novel Therapies for Type 1 Diabetes. Cells 2020; 9:cells9112403. [PMID: 33153010 PMCID: PMC7692667 DOI: 10.3390/cells9112403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune condition where the body’s immune cells destroy their insulin-producing pancreatic beta cells leading to dysregulated glycaemia. Individuals with T1D control their blood glucose through exogenous insulin replacement therapy, often using multiple daily injections or pumps. However, failure to accurately mimic intrinsic glucose regulation results in glucose fluctuations and long-term complications impacting key organs such as the heart, kidneys, and/or the eyes. It is well established that genetic and environmental factors contribute to the initiation and progression of T1D, but recent studies show that epigenetic modifications are also important. Here, we discuss key epigenetic modifications associated with T1D pathogenesis and discuss how recent research is finding ways to harness epigenetic mechanisms to prevent, reverse, or manage T1D.
Collapse
|
46
|
Li Y, Zhuang J. miR-345-3p serves a protective role during gestational diabetes mellitus by targeting BAK1. Exp Ther Med 2020; 21:2. [PMID: 33235611 PMCID: PMC7678625 DOI: 10.3892/etm.2020.9434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
Recent studies have demonstrated that microRNAs (miRs) serve a crucial role during the development of gestational diabetes mellitus (GDM). However, the mechanisms underlying miR-345-3p and its protective role during GDM have not been previously reported. The present study investigated miR-345-3p expression and function in vitro, and the possible molecular mechanisms underlying GDM. Compared with healthy pregnant women, miR-345-3p was downregulated in the placental tissue and peripheral blood of patients with GDM. Further investigation revealed that BCL2-antagonist/killer 1 (BAK1) was a predicted target gene of miR-345-3p, and the expression of BAK1 was significantly increased in patients with GDM compared with healthy pregnant women. In vitro analysis revealed that miR-345-3p mimic significantly increased cell viability, migration and invasion, inhibited apoptosis, upregulated Bcl-2 and matrix metallopeptidase 9 expression, and decreased Bax expression compared with the control group. Furthermore, miR-245-3p mimic-induced alterations were reversed by BAK1 overexpression. The results suggested that miR-345-3p overexpression exhibited a protective role in patients with GDM by inhibiting HTR8-/SVneo cell apoptosis, and promoting cell proliferation and migration via targeting BAK1. The use of miR-345-3p for the diagnosis of GDM requires further investigation.
Collapse
Affiliation(s)
- Yuxia Li
- Department of Gynecology and Obstetrics, Wuhan Children's Hospital, Wuhan Maternal and Child Healthcare Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430010, P.R. China
| | - Jun Zhuang
- Department of Obstetrics, Lianshui County People's Hospital, Huai'an, Jiangsu 223400, P.R. China
| |
Collapse
|
47
|
Saini J, Bandyopadhyay B, Pandey AD, Ramachandran VG, Das S, Sood V, Banerjee A, Vrati S. High-Throughput RNA Sequencing Analysis of Plasma Samples Reveals Circulating microRNA Signatures with Biomarker Potential in Dengue Disease Progression. mSystems 2020; 5:e00724-20. [PMID: 32934118 PMCID: PMC7498686 DOI: 10.1128/msystems.00724-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
The circulating microRNA (miRNA) profile has been widely used for identifying potential biomarkers against viral infections. However, data on circulating microRNA expression patterns in dengue patients are scanty. Considering the impact of severity caused by dengue infection, circulating miRNA profiles in plasma of dengue patients may prove to be valuable for developing early prognostic markers for the disease severity. Here, we described an in-depth analytical study of small RNA sequencing data obtained from the plasma of 39 dengue patients. Integrating bioinformatics and in vitro studies, we identified differentially expressed miRNAs (DEMs) (log2 fold change ≥1.5, P < 0.05) associated with dengue disease progression. In comparing miRNA expression pattern with the follow-up samples, nine miRNAs were found to exhibit an altered expression that could distinguish between severe dengue and the convalescent patients. To understand the abundance and specificity of the DEMs in the context of dengue infection and disease progression, eight top-hit DEMs were further validated in the dengue virus-infected cell lines as well as in the patient's plasma and peripheral blood mononuclear cells (PBMCs) using the quantitative reverse transcription-PCR (qRT-PCR) method. Importantly, receiver operating curve analysis further confirmed that the plasma expression pattern of hsa-miR-122-5p could differentiate between different stages of dengue infection (area under the concentration-time curve [AUC] = 0.792), and dengue-negative patients with other febrile illnesses (AUC = 0.984). The in silico analysis of DEM target genes suggested an enrichment of the pathways associated with metabolism and inflammation. Our study gives a global view of miRNA expression in the plasma from dengue patients and provides a precious resource of candidate miRNAs involved in dengue infection and disease progression.IMPORTANCE Dengue virus (DENV) infection usually causes dengue fever (DF) with flu-like illness affecting infants, young children, and adults. The DF occasionally evolves into a potentially lethal complication called dengue severe (DS) leading to a rapid fall in platelet count along with plasma leakage, fluid accumulation, respiratory distress, and severe bleeding. The diverse clinical spectrum of dengue disease, as well as its significant similarity to other febrile viral illnesses, makes early identification more challenging in this high-risk group. microRNAs (miRNAs) are small (∼19 to 21 nucleotides [nt] in length), noncoding RNAs, extremely stable and easily detectable in the plasma; thus, they have potential as biomarkers for diagnosing and monitoring human diseases. This study provides a comprehensive analysis of miRNAs circulating in plasma of dengue virus-infected patients and identifies the miRNA signatures that have biomarker potential for dengue infection and disease progression.
Collapse
Affiliation(s)
- Jaya Saini
- Regional Center for Biotechnology (RCB), Faridabad, India
| | | | - Abhay Deep Pandey
- Regional Center for Biotechnology (RCB), Faridabad, India
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - V G Ramachandran
- University College of Medical Sciences (UCMS) & Guru Teg Bahadur (GTB) Hospital, Delhi, India
| | - Shukla Das
- University College of Medical Sciences (UCMS) & Guru Teg Bahadur (GTB) Hospital, Delhi, India
| | - Vikas Sood
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
- Department of Biochemistry, Jamia Hamdard, New Delhi, India
| | - Arup Banerjee
- Regional Center for Biotechnology (RCB), Faridabad, India
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Sudhanshu Vrati
- Regional Center for Biotechnology (RCB), Faridabad, India
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
| |
Collapse
|
48
|
Chan HW, Yang B, Wong W, Blakeley P, Seah I, Tan QSW, Wang H, Bhargava M, Lin HA, Chai CHC, Mangunkusumo EA, Thet N, Yuen YS, Sethi R, Wang S, Hunziker W, Lingam G, Su X. A Pilot Study on MicroRNA Profile in Tear Fluid to Predict Response to Anti-VEGF Treatments for Diabetic Macular Edema. J Clin Med 2020; 9:E2920. [PMID: 32927780 PMCID: PMC7564365 DOI: 10.3390/jcm9092920] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
(1) Background: Intravitreal anti-vascular endothelial growth factor (anti-VEGF) is an established treatment for center-involving diabetic macular edema (ci-DME). However, the clinical response is heterogeneous. This study investigated miRNAs as a biomarker to predict treatment response to anti-VEGF in DME. (2) Methods: Tear fluid, aqueous, and blood were collected from patients with treatment-naïve DME for miRNA expression profiling with quantitative polymerase chain reaction. Differentially expressed miRNAs between good and poor responders were identified from tear fluid. Bioinformatics analysis with the miEAA tool, miRTarBase Annotations, Gene Ontology categories, KEGG, and miRWalk pathways identified interactions between enriched miRNAs and biological pathways. (3) Results: Of 24 participants, 28 eyes received bevacizumab (15 eyes) or aflibercept (13 eyes). Tear fluid had the most detectable miRNA species (N = 315), followed by serum (N = 309), then aqueous humor (N = 134). MiRNAs that correlated with change in macular thickness were miR-214-3p, miR-320d, and hsa-miR-874-3p in good responders; and miR-98-5p, miR-196b-5p, and miR-454-3p in poor responders. VEGF-related pathways and the angiogenin-PRI complex were enriched in good responders, while transforming growth factor-β and insulin-like growth factor pathways were enriched in poor responders. (4) Conclusions: We reported a panel of novel miRNAs that provide insight into biological pathways in DME. Validation in larger independent cohorts is needed to determine the predictive performance of these miRNA candidate biomarkers.
Collapse
Affiliation(s)
- Hwei Wuen Chan
- Department of Ophthalmology, National University Hospital, Singapore S118177, Singapore; (H.W.C.); (W.W.); (I.S.); (M.B.); (H.A.L.); (C.H.C.); (E.A.M.); (N.T.); (Y.S.Y.); (G.L.)
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (P.B.); (S.W.)
| | - Binxia Yang
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; (B.Y.); (Q.S.W.T.); (H.W.); (R.S.); (W.H.)
| | - Wendy Wong
- Department of Ophthalmology, National University Hospital, Singapore S118177, Singapore; (H.W.C.); (W.W.); (I.S.); (M.B.); (H.A.L.); (C.H.C.); (E.A.M.); (N.T.); (Y.S.Y.); (G.L.)
| | - Paul Blakeley
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (P.B.); (S.W.)
| | - Ivan Seah
- Department of Ophthalmology, National University Hospital, Singapore S118177, Singapore; (H.W.C.); (W.W.); (I.S.); (M.B.); (H.A.L.); (C.H.C.); (E.A.M.); (N.T.); (Y.S.Y.); (G.L.)
| | - Queenie Shu Woon Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; (B.Y.); (Q.S.W.T.); (H.W.); (R.S.); (W.H.)
| | - Haofei Wang
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; (B.Y.); (Q.S.W.T.); (H.W.); (R.S.); (W.H.)
| | - Mayuri Bhargava
- Department of Ophthalmology, National University Hospital, Singapore S118177, Singapore; (H.W.C.); (W.W.); (I.S.); (M.B.); (H.A.L.); (C.H.C.); (E.A.M.); (N.T.); (Y.S.Y.); (G.L.)
| | - Hazel Anne Lin
- Department of Ophthalmology, National University Hospital, Singapore S118177, Singapore; (H.W.C.); (W.W.); (I.S.); (M.B.); (H.A.L.); (C.H.C.); (E.A.M.); (N.T.); (Y.S.Y.); (G.L.)
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (P.B.); (S.W.)
| | - Charmaine HC Chai
- Department of Ophthalmology, National University Hospital, Singapore S118177, Singapore; (H.W.C.); (W.W.); (I.S.); (M.B.); (H.A.L.); (C.H.C.); (E.A.M.); (N.T.); (Y.S.Y.); (G.L.)
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (P.B.); (S.W.)
| | - Erlangga Ariadarma Mangunkusumo
- Department of Ophthalmology, National University Hospital, Singapore S118177, Singapore; (H.W.C.); (W.W.); (I.S.); (M.B.); (H.A.L.); (C.H.C.); (E.A.M.); (N.T.); (Y.S.Y.); (G.L.)
| | - Naing Thet
- Department of Ophthalmology, National University Hospital, Singapore S118177, Singapore; (H.W.C.); (W.W.); (I.S.); (M.B.); (H.A.L.); (C.H.C.); (E.A.M.); (N.T.); (Y.S.Y.); (G.L.)
| | - Yew Sen Yuen
- Department of Ophthalmology, National University Hospital, Singapore S118177, Singapore; (H.W.C.); (W.W.); (I.S.); (M.B.); (H.A.L.); (C.H.C.); (E.A.M.); (N.T.); (Y.S.Y.); (G.L.)
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (P.B.); (S.W.)
| | - Raman Sethi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; (B.Y.); (Q.S.W.T.); (H.W.); (R.S.); (W.H.)
| | - Si Wang
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (P.B.); (S.W.)
| | - Walter Hunziker
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; (B.Y.); (Q.S.W.T.); (H.W.); (R.S.); (W.H.)
| | - Gopal Lingam
- Department of Ophthalmology, National University Hospital, Singapore S118177, Singapore; (H.W.C.); (W.W.); (I.S.); (M.B.); (H.A.L.); (C.H.C.); (E.A.M.); (N.T.); (Y.S.Y.); (G.L.)
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (P.B.); (S.W.)
| | - Xinyi Su
- Department of Ophthalmology, National University Hospital, Singapore S118177, Singapore; (H.W.C.); (W.W.); (I.S.); (M.B.); (H.A.L.); (C.H.C.); (E.A.M.); (N.T.); (Y.S.Y.); (G.L.)
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (P.B.); (S.W.)
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; (B.Y.); (Q.S.W.T.); (H.W.); (R.S.); (W.H.)
- Singapore Eye Research Institute (SERI), Singapore National Eye Centre, Singapore 169856, Singapore
| |
Collapse
|
49
|
Liu Y, Xu J, Gu R, Li Z, Wang K, Qi Y, Sun X, Xie J, Wang L, Xu B, Kang L. Circulating exosomal miR-144-3p inhibits the mobilization of endothelial progenitor cells post myocardial infarction via regulating the MMP9 pathway. Aging (Albany NY) 2020; 12:16294-16303. [PMID: 32843584 PMCID: PMC7485705 DOI: 10.18632/aging.103651] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/19/2020] [Indexed: 12/15/2022]
Abstract
Background: The angiogenesis post myocardial infarction (MI) is compromised in diabetes. MiR-144-3p is reported to be highly expressed in circulating exosomes of diabetic patients, implying its role in diabetic complications. However, whether circulating exosomes and enriched miR-144-3p are involved in the impaired neovascularization in diabetes and the underlying mechanism is unclear. Results: DMexo and miR-144-3p mimic-treated MSCs had elevated miR-144-3p levels and decreased MMP9, Ets1 and PLG expression. The percentage of EPCs were relatively lower in DMexo-treated or agomir-treated MI mice compared with MI mice. Finally, the luciferase assay confirmed the direct binding between miR-144-3p and Ets1. Conclusion: Exosomal miR-144-3p could impair the mobilization ability of EPCs, which was associated with impaired ischemia-induced neovascularization. Methods: Circulating exosomes were isolated from Streptozotocin (STZ)-induced mice. In vitro, mesenchymal stem cells (MSCs) were incubated with exosomes from diabetic mice (DMexo), and miR-144-3p mimic or inhibitor. miR-144-3p, and MMP9 pathway were measured using qPCR and immunoblotting. In vivo, MI mice induced by left anterior descending ligation were treated with DMexo, as well as miR-144-3p agomir. Flow cytometry was used to profile endothelial progenitor cells (EPCs) in peripheral blood and bone marrow post 24 hours respectively.
Collapse
Affiliation(s)
- Yihai Liu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Jiamin Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Rong Gu
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Zhu Li
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Kun Wang
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Yu Qi
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Xuan Sun
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Jun Xie
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Lian Wang
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China.,Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Lina Kang
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China.,Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Emerging data have suggested that β-cell dysfunction may exacerbate the development and progression of type 1 diabetes (T1D). In this review, we highlight clinical and preclinical studies suggesting a role for β-cell dysfunction during the evolution of T1D and suggest agents that may promote β-cell health in T1D. RECENT FINDINGS Metabolic abnormalities exist years before development of hyperglycemia and exhibit a reproducible pattern reflecting progressive deterioration of β-cell function and increases in β-cell stress and death. Preclinical studies indicate that T1D may be prevented by modification of pathways impacting intrinsic β-cell stress and antigen presentation. Recent findings suggest that differences in metabolic phenotypes and β-cell stress may reflect differing endotypes of T1D. Multiple pathways representing potential drug targets have been identified, but most remain to be tested in human populations with preclinical disease. SUMMARY This cumulative body of work shows clear evidence that β-cell stress, dysfunction, and death are harbingers of impending T1D and likely contribute to progression of disease and insulin deficiency. Treatment with agents targeting β-cell health could augment interventions with immunomodulatory therapies but will need to be tested in intervention studies with endpoints carefully designed to capture changes in β-cell function and health.
Collapse
Affiliation(s)
- Emily K. Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Department of Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Department of Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center and the Department of Medicine, The University of Chicago, Chicago, IL
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Department of Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Department of Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Roudebush VA Medical Center, Indianapolis, IN
| |
Collapse
|