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
|
Shnayder NA, Pekarets NA, Pekarets NI, Dmitrenko DV, Grechkina VV, Petrova MM, Al-Zamil M, Nasyrova RF. MicroRNAs as Epigenetic Biomarkers of Pathogenetic Mechanisms of the Metabolic Syndrome Induced by Antiseizure Medications: Systematic Review. J Clin Med 2025; 14:2432. [PMID: 40217882 PMCID: PMC11989458 DOI: 10.3390/jcm14072432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/27/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Antiseizure medication (ASM) induced metabolic syndrome (AIMetS) is a common adverse drug reaction (ADR) of pharmacotherapy for epilepsy and psychiatric disorders. However, the sensitivity and specificity of blood biomarkers may be insufficient due to the influence of combined pathology, concomitant diseases, and the peculiarities of the metabolism of ASMs in patients with epilepsy. Methods: The presented results of experimental and clinical studies of microRNAs (miRs) as epigenetic biomarkers of MetS and AIMetS, which were entered into the different databases, were analyzed for the last decade (2014-2024). Results: A systematic review demonstrated that miRs can act as promising epigenetic biomarkers of key AIMetS domains. However, the results of the review demonstrated the variable role of various miRs and their paralogs in the pathogenesis of AIMetS. Therefore, as part of this study, an miRs signature was proposed that allows us to assess the risk of developing and the severity of AIMetS as low risk, medium risk, and high risk. Conclusions: The mechanisms of development and biomarkers of AIMetS are an actual problem of epileptology, which is still far from being resolved. The development of panels (signatures) of epigenetic biomarkers of this widespread ADR may help to increase the safety of pharmacotherapy of epilepsy. However, to increase the sensitivity and specificity of circulating miRs in the blood as biomarkers of AIMetS, it is necessary to conduct "bridge" studies in order to replicate the results of preclinical and clinical studies into real clinical practice.
Collapse
Affiliation(s)
- Natalia A. Shnayder
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 1 Partizan Zheleznyak St., 660022 Krasnoyarsk, Russia; (D.V.D.); (M.M.P.)
| | - Nikolai A. Pekarets
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
| | - Natalia I. Pekarets
- Department of Psychiatry and Clinical Psychology, Irkutsk State Medical University, 1 Krasny Vosstaniya St., 664003 Irkutsk, Russia;
| | - Diana V. Dmitrenko
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 1 Partizan Zheleznyak St., 660022 Krasnoyarsk, Russia; (D.V.D.); (M.M.P.)
| | - Violetta V. Grechkina
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
| | - Marina M. Petrova
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 1 Partizan Zheleznyak St., 660022 Krasnoyarsk, Russia; (D.V.D.); (M.M.P.)
| | - Mustafa Al-Zamil
- Department of Physiotherapy, Faculty of Continuing Medical Education, Peoples’ Friendship University of Russia, 117198 Moscow, Russia;
| | - Regina F. Nasyrova
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
- Department of Psychiatry, General and Clinical Psychology, Tula State University, 92 Lenin Ave., 300012 Tula, Russia
| |
Collapse
|
3
|
D'Italia G, Schroen B, Cosemans JM. Commonalities of platelet dysfunction in heart failure with preserved ejection fraction and underlying comorbidities. ESC Heart Fail 2025; 12:1013-1028. [PMID: 39375979 PMCID: PMC11911585 DOI: 10.1002/ehf2.15090] [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: 06/10/2024] [Revised: 08/06/2024] [Accepted: 09/06/2024] [Indexed: 10/09/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by a lack of a specific targeted treatment and a complex, partially unexplored pathophysiology. Common comorbidities associated with HFpEF are hypertension, atrial fibrillation, obesity and diabetes. These comorbidities, combined with advanced age, play a crucial role in the initiation and development of the disease through the promotion of systemic inflammation and consequent changes in cardiac phenotype. In this context, we suggest platelets as important players due to their emerging role in vascular inflammation. This review provides an overview of the role of platelets in HFpEF and its associated comorbidities, including hypertension, atrial fibrillation, obesity and diabetes mellitus, as well as the impact of age and sex on platelet function. These major HFpEF-associated comorbidities present alterations in platelet behaviour and in features linked to platelet size, content and reactivity. The resulting dysfunctional platelets can contribute to further increase inflammation, oxidative stress and endothelial dysfunction, suggesting an active role of these cells in the initiation and progression of HFpEF. Recent evidence shows that reduced platelet count and elevated mean platelet volume are associated with worsening heart failure in HFpEF patients. However, the specific mechanisms by which platelets contribute to HFpEF development and progression are still largely unexplored, with only a few studies investigating platelet function in HFpEF. We discuss the limited yet significant body of research investigating platelet function in HFpEF, emphasizing the need for more comprehensive studies. Additionally, we explore the potential mechanisms through which platelets may influence HFpEF, such as their interactions with the vascular endothelium and the secretion of bioactive molecules like cytokines, chemokines and RNA molecules. These interactions and secretions may play a role in modulating vascular inflammation and contributing to the pathophysiological landscape of HFpEF. The review underscores the necessity for future research to elucidate the precise contributions of platelets to HFpEF, aiming to potentially identify novel therapeutic targets and improve patient outcomes. The evidence presented herein supports the hypothesis that platelets are not merely passive bystanders but active participants in the pathophysiology of HFpEF and its comorbidities.
Collapse
Affiliation(s)
- Giorgia D'Italia
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Blanche Schroen
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Judith M.E.M. Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
4
|
Mohamed Abdelgawwad El-Sehrawy AA, Mohammed MH, Salahldin OD, Uthirapathy S, Ballal S, Kalia R, Arya R, Joshi KK, Kadim AS, Kadhim AJ. Crosstalk between microRNA and inflammation; critical regulator of diabetes. Exp Cell Res 2025; 447:114507. [PMID: 40058448 DOI: 10.1016/j.yexcr.2025.114507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
A growing body of evidence indicates that microRNAs (miRNAs may be used as biomarkers for the diagnosis, prognosis, and treatment of diabetes, given their changed expression profile as the disease progresses. There is growing interest in using individual miRNAs or whole miRNA clusters linked to diabetes as therapeutic targets because of their abnormal expression and functioning. In diabetes, miRNAs are also involved in inflammatory and immunological responses. Additionally, the inflammatory response controls the generation, processing, and stability of pre- or mature miRNAs and miRNA biogenesis. With a comprehensive grasp of molecular biological activities and the signaling axis, this review emphasizes the critical functions of miRNAs in inflammatory and immunological processes in diabetes. We further emphasized the potential role of these miRNAs in controlling inflammation associated with diabetes. This assessment will direct the shift from many studies to practical applications for tailored diabetes treatment and assist in identifying new therapeutic targets and approaches.
Collapse
Affiliation(s)
| | - Mohammed Hashim Mohammed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, Al-maarif University, Anbar, Iraq.
| | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India.
| | - Rishiv Kalia
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Renu Arya
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India.
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, 248002, Uttarakhand, India; Graphic Era Deemed to Be University, Dehradun, Uttarakhand, India.
| | - Arshed Shakir Kadim
- Radiological Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, Babylon, 51001, Iraq.
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq.
| |
Collapse
|
5
|
Bagni G, Biancalana E, Chiara E, Costanzo I, Malandrino D, Lastraioli E, Palmerini M, Silvestri E, Urban ML, Emmi G. Epigenetics in autoimmune diseases: Unraveling the hidden regulators of immune dysregulation. Autoimmun Rev 2025; 24:103784. [PMID: 40043893 DOI: 10.1016/j.autrev.2025.103784] [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: 10/31/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/09/2025]
Abstract
Autoimmune diseases result from complex interactions between genetic and environmental factors. Recent advances in epigenetic research shed light on the intricate regulatory mechanisms that contribute to the development and progression of such conditions. The present review aims to explore the role of epigenetic modifications, including DNA methylation, histone modifications, and non-coding RNAs, in the context of autoimmune diseases. We discuss the current understanding of epigenetic alterations associated with various autoimmune disorders, their impact on immune cell function, and their potential as innovative therapeutic targets. Additionally, we highlight the main future directions in the field of epigenetics in autoimmunity.
Collapse
Affiliation(s)
- Giacomo Bagni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Edoardo Biancalana
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Emanuele Chiara
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Iole Costanzo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Danilo Malandrino
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Elena Lastraioli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Miki Palmerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elena Silvestri
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Maria Letizia Urban
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Giacomo Emmi
- Department of Medical, Surgery and Health Sciences, University of Trieste, and Clinical Medicine and Rheumatology Unit, Cattinara University Hospital, Trieste, Italy; Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Melbourne, Australia.
| |
Collapse
|
6
|
Li T, Xu L, Shen P, Qiu J, Wang Y, Hu J, Guan P, Lin H, Jiang Z, Chen K, Wang J. The role of miRNAs in the associations between particulate matter and ischemic stroke: A nested case-control study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125750. [PMID: 39870131 DOI: 10.1016/j.envpol.2025.125750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/03/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025]
Abstract
Epidemiological studies have reported that atmospheric particulate matter (PM) contributes to ischemic stroke (IS). Biological studies also indicated that the pathway where PM induces IS involves several pathological processes. Moreover, exposure to PM can alter the expression of specific microRNAs (miRNAs) and ultimately accelerate the onset of IS by regulating related pathways. However, evidence on the role of miRNAs between PM and IS still needs to be fully elucidated. We used the miRNA sequencing datasets from the GEO (Gene Expression Omnibus) to screen miRNAs associated with IS. A nested case-control study was performed, including all incident ischemic stroke cases during the follow-up period and controls matched by age, sex, and entry seasons. Land use regression (LUR) models were constructed to estimate the levels of PM2.5 and PM10. The real-time quantitative PCR (RT-qPCR) assay was applied to detect the expression of candidate miRNAs in plasma samples collected at baseline to verify whether candidate miRNAs differentially expressed between cases and controls. Mediation analyses were applied to evaluate whether PM could induce IS by affecting the expression of miRNAs. We screened 23 miRNAs expressed differentially between cases and controls from the GEO database. A total of 605 incident ischemic stroke patients were finally included in the case group, and 605 healthy controls were matched. The RT-qPCR assay detected 15 differentially expressed miRNAs. Mediating effects of hsa-miR-107, hsa-miR-320b, hsa-miR-423-5p, hsa-miR-483-5p, and hsa-miR-935 were observed for the associations between PM and IS, indicating that PM could promote IS by altering the expression of those miRNAs. In this nested case-control study, PM might induce IS by affecting the expression of hsa-miR-107, hsa-miR-320b, hsa-miR-423-5p, hsa-miR-483-5p and hsa-miR-935.
Collapse
Affiliation(s)
- Tiezheng Li
- Department of Public Health, and Department of Endocrinology of the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, China
| | - Lisha Xu
- Department of Public Health, and Department of Endocrinology of the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, China
| | - Peng Shen
- Department of Chronic Disease and Health Promotion, Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Jie Qiu
- Department of Public Health, and Department of Endocrinology of the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, China
| | - Yixing Wang
- Department of Public Health, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Hu
- Department of Public Health, and Department of Endocrinology of the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, China
| | - Peng Guan
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Hongbo Lin
- Department of Chronic Disease and Health Promotion, Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Zhiqin Jiang
- Department of Chronic Disease and Health Promotion, Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Kun Chen
- Department of Public Health, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jianbing Wang
- Department of Public Health, and Department of Endocrinology of the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, China.
| |
Collapse
|
7
|
Wang Y, Liu S, Zhou Q, Feng Y, Xu Q, Luo L, Lv H. Bioinformatics for the Identification of STING-Related Genes in Diabetic Retinopathy. Curr Eye Res 2025; 50:320-333. [PMID: 39704112 DOI: 10.1080/02713683.2024.2430223] [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: 04/13/2024] [Revised: 09/06/2024] [Accepted: 11/11/2024] [Indexed: 12/21/2024]
Abstract
PURPOSE Diabetic retinopathy (DR) is the most common complication of diabetes mellitus. Stimulator of interferon genes (STING) plays an important regulatory role in the transcription of several genes. This study aimed to mine and identify hub genes relevant to STING in DR. METHODS The STING-related genes (STING-RGs) were extracted from MSigDB database. Differentially expressed STING-RGs (DE-STING-RGs) were filtered by overlapping differentially expressed genes (DEGs) between DR and NC specimens and STING-RGs. A PPI network was established to mine hub genes. The ability of the hub genes to differentiate between DR and NC specimens was evaluated. Additionally, a ceRNA network was established to investigate the regulatory mechanisms of hub genes. Subsequently, the discrepancies in immune infiltration between DR and NC specimens were further explored. Additionally, we performed drug predictions. Finally, RT-qPCR of peripheral blood samples was used to validate the bioinformatics results. RESULTS A grand total of four genes (IKBKG, STAT6, NFKBIA, and FCGR2A) related to STING were identified for DR. The AUC values of all four hub genes were greater than 0.7, which indicated that the diagnostic value was acceptable. The ceRNA network contained four hub genes, 170 miRNAs, and 135 lncRNAs. In addition, immunoinfiltration analysis demonstrated that the abundance of activated B cells was notably different between the DR and NC specimens. Moreover, 32 drugs were included in the drug-gene network, with twelve drugs targeting STAT6, nine drugs targeting NFKBIA, four drugs targeted IKBKG, and seven drugs targeted FCGR2A. The expression of the four hub genes in blood samples determined by RT-qPCR was consistent with our analysis. CONCLUSION In conclusion, four hub genes (IKBKG, STAT6, NFKBIA, and FCGR2A) related to STING with a diagnostic value for DR were identified by bioinformatics analysis, which might provide new insights into the evaluation and treatment of DR.
Collapse
Affiliation(s)
- Yu Wang
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Siyan Liu
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Qi Zhou
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Yalin Feng
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Qin Xu
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Linbi Luo
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Hongbin Lv
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| |
Collapse
|
8
|
Castoldi M, Roy S, Angendohr C, Pellegrino R, Vucur M, Singer MT, Buettner V, Dille MA, Wolf SD, Heij LR, Ghallab A, Albrecht W, Hengstler JG, Flügen G, Knoefel WT, Bode JG, Zender L, Neumann UP, Heikenwälder M, Longerich T, Roderburg C, Luedde T. Regulation of KIF23 by miR-107 controls replicative tumor cell fitness in mouse and human hepatocellular carcinoma. J Hepatol 2025; 82:499-511. [PMID: 40235270 DOI: 10.1016/j.jhep.2024.08.025] [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: 12/09/2023] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 04/17/2025]
Abstract
BACKGROUND & AIMS In hepatocellular carcinoma (HCC), successful translation of experimental targets identified in mouse models to human patients has proven challenging. In this study, we used a comprehensive transcriptomic approach in mice to identify novel potential targets for therapeutic intervention in humans. METHODS We analyzed combined genome-wide miRNA and mRNA expression data in three pathogenically distinct mouse models of liver cancer. Effects of target genes on hepatoma cell fitness were evaluated by proliferation, survival and motility assays. TCGA and GEO databases, in combination with tissue microarrays, were used to validate the mouse targets and their impact on human HCC prognosis. Finally, the functional effects of the identified targets on tumorigenesis and tumor therapy were tested in hydrodynamic tail vein injection-based preclinical HCC models in vivo. RESULTS The expression of miR-107 was found to be significantly reduced in mouse models of liver tumors of various etiologies and in cohorts of humans with HCC. Overexpression of miR-107 or inhibition of its novel target kinesin family member 23 (Kif23) significantly reduced proliferation by interfering with cytokinesis, thereby controlling survival and motility of mouse and human hepatoma cells. In humans, KIF23 expression was found to be a prognostic marker in liver cancer, with high expression associated with poor prognosis. Hydrodynamic tail vein injection of vectors carrying either pre-miR-107 or anti-Kif23 shRNA inhibited the development of highly aggressive c-Myc-NRAS-induced liver cancers in mice. CONCLUSIONS Disruption of the miR-107/Kif23 axis inhibited hepatoma cell proliferation in vitro and prevented oncogene-induced liver cancer development in vivo, offering a novel potential avenue for the treatment of HCC in humans. IMPACT AND IMPLICATIONS Our study revealed the central role of the miR-107/KIF23 axis in controlling tumor cell fitness and hepatocellular carcinoma progression. The results demonstrate that the overexpression of miR-107 or silencing of its target, KIF23, markedly suppresses the proliferation, survival, and motility of human and mouse hepatoma cells. In this work, we demonstrate that the disruption of miR-107/Kif23 signaling effectively protects mice from an aggressive form of oncogene-induced liver cancer in vivo, implying that targeting miR-107/KIF23 might be a novel therapeutic approach for hepatocellular carcinoma in humans.
Collapse
Affiliation(s)
- Mirco Castoldi
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany.
| | - Sanchari Roy
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Carolin Angendohr
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Rossella Pellegrino
- Institute of Pathology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Mihael Vucur
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Michael T Singer
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Veronika Buettner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Matthias A Dille
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Stephanie D Wolf
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Lara R Heij
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany; Department of Surgery and Transplantation, University Hospital Essen, Essen, Germany; Department of Pathology, Erasmus Medical Center Rotterdam, The Netherlands
| | - Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, Dortmund, Germany; Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Wiebke Albrecht
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, Dortmund, Germany
| | - Georg Flügen
- Department of Surgery and Transplantation, University Hospital Essen, Essen, Germany; Department of Surgery, Heinrich-Heine-University and University Hospital Düsseldorf, Germany
| | - Wolfram T Knoefel
- Department of Surgery, Heinrich-Heine-University and University Hospital Düsseldorf, Germany
| | - Johannes G Bode
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Lars Zender
- Department of Internal Medicine VIII, University Hospital Tubingen, Tubingen, Germany
| | - Ulf P Neumann
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany; Department of Surgery and Transplantation, University Hospital Essen, Essen, Germany
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Longerich
- Institute of Pathology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany.
| |
Collapse
|
9
|
Lin YK, Pan YF, Jiang TY, Chen YB, Shang TY, Xu MY, Feng HB, Ma YH, Tan YX, Wang HY, Dong LW. Blocking the SIRPα-CD47 axis promotes macrophage phagocytosis of exosomes derived from visceral adipose tissue and improves inflammation and metabolism in mice. J Biomed Sci 2025; 32:31. [PMID: 40016734 PMCID: PMC11869713 DOI: 10.1186/s12929-025-01124-y] [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: 10/24/2024] [Accepted: 02/06/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Adipose tissue plays a pivotal role in systemic metabolism and maintaining bodily homeostasis. Exosomes from adipose tissues, known as AT-Exos, are recognized as important messengers in the communication between adipose tissue and other organs. Despite this, the alterations in exosome composition and the functional disparities among depot-specific AT-Exos in obesity remain elusive. METHODS In this work, we utilized lipidomics and microRNA (miRNA) sequencing to elucidate the lipid and miRNA profiles of AT-Exos in a diet-induced obesity model. We identified obesity-related miRNAs in AT-Exos and further explored their mechanisms using gain- and loss-of-function experiments. To evaluate the metabolic effects of AT-Exos on adipocytes, we conducted RNA-sequencing (RNA-seq) and confirmed our findings through Quantitative Real-time PCR (qPCR) and Western bolt analyses. Meanwhile, a mouse model with intraperitoneal injections was utilized to validate the role of exosomes derived from visceral white adipose tissue (vWAT-Exos) in obesity progression in vivo. Finally, we explored potential therapeutic intervention strategies targeting AT-Exos, particularly focusing on modulating the SIRPα-CD47 axis to enhance macrophage phagocytosis using Leptin-deficient (ob/ob) mice and SIRPα knock-out mice. RESULTS Our study revealed that obesity-related metabolism affects the biological processes of AT-Exos, with depot-specific secretion patterns. In obesity, the lipidome profile of AT-Exos was significantly altered, and diet can modify the miRNA content and function within these exosomes, influencing lipid metabolism and inflammatory pathways that contribute to metabolic dysregulation. Specifically, we identified that miR-200a-3p and miR-200b-3p promoted lipid accumulation in 3T3L1 cells partly through the PI3K/AKT/mTOR pathway. RNA-Seq analysis revealed that AT-Exos from different fat depots exerted distinct effects on adipocyte metabolism, with obese vWAT-Exos being notably potent in triggering inflammation and lipid accumulation in diet-induced obesity. Additionally, we found that inhibiting the SIRPα-CD47 axis can mitigate metabolic disorders induced by obese vWAT-Exos or ob/ob mice, partly due to the enhanced clearance of vWAT-Exos. Consistent with this, SIRPα-deficient mice exhibited a reduction in vWAT-Exos and displayed greater resistance to obesity. CONCLUSIONS This study elucidates that diet-induced obesity altered the lipid and miRNA profiles of AT-Exos, which involved in modulating adipocyte inflammation and metabolic balance. The SIRPα-CD47 axis emerges as a potential therapeutic target for obesity and its associated complications.
Collapse
Affiliation(s)
- Yun-Kai Lin
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Yu-Fei Pan
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Tian-Yi Jiang
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Yi-Bin Chen
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Tai-Yu Shang
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Meng-You Xu
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Hui-Bo Feng
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Yun-Han Ma
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Ye-Xiong Tan
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Hong-Yang Wang
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Li-Wei Dong
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China.
| |
Collapse
|
10
|
Shao B, Wang Z, Luo P, Du P, Zhang X, Zhang H, Si X, Ma S, Chen W, Huang Y. Identifying insulin-responsive circRNAs in chicken pectoralis. BMC Genomics 2025; 26:148. [PMID: 39955508 PMCID: PMC11830218 DOI: 10.1186/s12864-025-11347-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) are stable, covalently closed non-coding RNAs formed by reverse splicing of precursor mRNA. They play critical roles in various biological processes, including insulin secretion and metabolism. However, their function in avian skeletal muscle's response to insulin remains poorly understood. This study aimed to comprehensively identify insulin-responsive circRNAs and explore their temporal and breed-specific regulation in poultry. RESULTS Using strand-specific RNA sequencing (ssRNA-Seq) on the pectoralis muscles of both Arbor Acres (AA) broilers and Silky fowls following insulin administration (5 IU/kg.BW, PBS as control). We identified 2,027 muscle circRNAs. Insulin-responsive circRNAs were detected in Silky fowls (29) and broilers (45) at 120 min, and in broilers (20) at 15 min post-injection. These circRNAs are enriched in processes such as exocrine pancreas development, response to exogenous stimuli, and regulation of intracellular signal transduction, likely mediated through a circRNA-miRNA network. Fewer insulin-responsive circRNAs were shared between time points in broilers (1) or between breeds (3) at 120 min. We further characterized a conserved insulin-responsive circRNA (circINSR), formed by exon 2 of the Insulin Receptor (INSR). The circINSR showed a similar spatiotemporal expression pattern to INSR, but exhibited distinct changes post-insulin administration. In broilers, INSR expression was dynamically modulated, while circINSR was downregulated only at 15 min (P < 0.01). Conversely, glucose did not change muscle circINSR but increased INSR at 10 min (P < 0.01). Energy restriction significantly downregulated circINSR (P < 0.01) without affecting INSR levels, and pyruvate had no effect on either circINSR or INSR levels. CONCLUSION This study reveals the dynamic and breed-specific roles of circRNAs, particularly circINSR, in avian skeletal muscle's response to insulin. The distinct regulation of circINSR and INSR under various metabolic conditions suggests a complex regulatory mechanism. These findings provide novel insights into the molecular basis of insulin signaling in avian species and highlight the potential of circRNAs as biomarkers for metabolic regulation.
Collapse
Affiliation(s)
- Binghao Shao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Ziyang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Pengna Luo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Pengfei Du
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xiangli Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Huaiyong Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xuemeng Si
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Sen Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Wen Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China.
| | - Yanqun Huang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China.
| |
Collapse
|
11
|
Milan KL, Anuradha M, Ramkumar KM. Role of miR-125b-5p in modulating placental SIRT7 expression and its implications for lipid metabolism in gestational diabetes. J Reprod Immunol 2025; 167:104422. [PMID: 39755065 DOI: 10.1016/j.jri.2024.104422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025]
Abstract
Gestational diabetes is marked impaired glucose tolerance, poses various adverse outcomes including increased BMI and obesity. These outcomes results from excess lipid accumulation which is marked by elevated triglycerides. In GDM, placenta exhibits altered lipid metabolism, including reduced fatty acid oxidation and increased triglyceride accumulation. These elevated triglycerides can also contribute to oxidative stress in GDM. SIRT7 plays an important role in regulating lipid metabolism and triglycerides levels. This study aimed to investigate the potential of miRNA to regulate the placental SIRT7 in GDM. PCR analysis reveals that SIRT7 expression along with oxidative stress markers elevated in GDM placenta. These elevated SIRT7 levels were positively correlated with BMI and triglycerides levels in GDM subjects. miR-125b-5p was identified to regulate SIRT7 mRNA using in-silico approaches. Expression levels of miR-125b-5p were found to be downregulated in GDM placenta and found to be negatively correlated with SIRT7 mRNA expression. To confirm the hypothesis BeWo cells were transfected with anti-miR-125b and miR-125b-mimic. Anti-miR overexpressed the SIRT7 expression where mimic dysregulated it. Additionally, overexpressing miR-125b-5p controlled the elevated SIRT7 caused by the exposure of high glucose in BeWo cells. Collectively this study indicated that miR-125b-5p may regulate lipid metabolism via SIRT7 contributing to GDM. These findings highlights the warrant of further research to develop the therapeutic approaches that target miR-125b-5p to reduce lipid accumulation and obesity in GDM.
Collapse
Affiliation(s)
- K L Milan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - M Anuradha
- Department of Obstetrics & Gynaecology, SRM Medical College Hospital and Research Centre, Kattankulathur, Tamil Nadu 603203, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India.
| |
Collapse
|
12
|
Jia X, Liu J, Jiang W, Chang L, Shen X, Jiang G, Li X, Chi C, Liu W, Zhang D. Binding site redundancy is critical for the regulation of fas by miR-30c in blunt snout bream (Megalobrama amblycephala). Comp Biochem Physiol A Mol Integr Physiol 2025; 299:111763. [PMID: 39395751 DOI: 10.1016/j.cbpa.2024.111763] [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: 09/11/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
MiR-30c and fatty acid synthase (fas) both play important roles in physiological processes such as lipid synthesis and fat metabolism. Predictive analysis revealed that fas is a target gene of miR-30c with multiple seed sites. Seed sites are useful to predict miRNA targeting relationships; however, detailed analyses of seed sites in fish genomes remain poorly studied. In this study, the regulatory relationship between miR-30c and fas, number and effect of seed regions, and mechanism by which miR-30c regulates lipid metabolism were evaluated in blunt snout bream (Megalobrama amblycephala). Four miR-30c target sites for fas were identified using various prediction tools. miR-30c mimics were transfected into 293 T cells, and dual-luciferase reporter assays were used to evaluate the roles of different fas target sites. When a single target site was mutated, relative luciferase activity was higher than that in the control group, with different activity levels depending on the mutation site. When multiple target sites were mutated, relative luciferase activity increased significantly as the number of mutation sites increased and was the highest when the four sites were mutated simultaneously. The miR-30c agomir was injected into the abdominal cavity of M. amblycephala at various concentrations for analyses of physiological and biochemical parameters in the liver and blood and the expression of genes related to lipid metabolism in the liver. Total cholesterol, free fatty acid, triglyceride, and low density lipoprotein levels were significantly lower after miR-30c agomir injection comparing to the control (P < 0.05). Additionally, the expression levels of genes related to lipid metabolism were significantly lower after miR-30c agomir injection than in the control (P < 0.05). In summary, this study identified four specific miR-30c target sites in the 3' UTR of fas mRNA; the effects of these sites are cumulative, and the redundancy ensures the accurate regulation of fas during evolution. In addition, miR-30c has a negative regulatory effect on fas and regulates lipid metabolism via various genes related to this process. Therefore, the regulation of miR-30c can effectively ameliorate the side effects of a high-fat diet on liver function in M. amblycephala.
Collapse
Affiliation(s)
- Xiaoyan Jia
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jie Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weibo Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Le Chang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoxue Shen
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Guangzhen Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangfei Li
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Cheng Chi
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wenbin Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Dingdong Zhang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
13
|
Patterson RA, Cho NA, Fernandes TS, Tuplin EWN, Lowry DE, Silva GAV, Reimer RA. Effects of a paternal diet high in animal protein (casein) versus plant protein (pea protein with added methionine) on offspring metabolic and gut microbiota outcomes in rats. Appl Physiol Nutr Metab 2025; 50:1-15. [PMID: 39689296 DOI: 10.1139/apnm-2024-0294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Evidence suggests that paternal diet can influence offspring metabolic health intergenerationally but whether dietary animal and plant proteins differ in their impact on fathers and their offspring is not known. Our objective was to examine the effects of a paternal diet high in casein versus pea protein on fathers and their offspring. Five-week-old male rats were fed: (1) control, (2) high animal protein (AP, 36.1% of energy as casein), or (3) high plant protein (PP, 36.1% of energy as pea protein with added methionine) diets for 8-11 weeks before being mated. Offspring were challenged with a high fat/sucrose diet (HFD) from 10 to 16 weeks of age. Metabolic and microbial outcomes were assessed in both generations. In fathers fed PP diet, enhanced insulin sensitivity and lower liver triglycerides were seen alongside altered hepatic microRNA expression and gut microbial profiles. Few changes were seen in their offspring. In contrast, the paternal AP diet influenced adult offspring hepatic microRNA expression and programmed a latent increase in adiposity, dysregulated satiety hormones, and modified gut microbial composition in their adult offspring that occurred following the HFD. Overall, a diet high in pea protein with added methionine demonstrated protective effects on biomarkers of metabolic health in the fathers but led to minimal effects on the offspring while a paternal diet high in casein led to evidence of an increase in characteristics of metabolic dysfunction in their adult offspring when unmasked by exposure to a HFD for 6 weeks.
Collapse
Affiliation(s)
- Riley A Patterson
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Nicole A Cho
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Tyra S Fernandes
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Erin W Noye Tuplin
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Dana E Lowry
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Gabriel A Venegas Silva
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
14
|
Darwish R, Alcibahy Y, Bucheeri S, Albishtawi A, Tama M, Shetty J, Butler AE. The Role of Hypothalamic Microglia in the Onset of Insulin Resistance and Type 2 Diabetes: A Neuro-Immune Perspective. Int J Mol Sci 2024; 25:13169. [PMID: 39684879 DOI: 10.3390/ijms252313169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Historically, microglial activation has been associated with diseases of a neurodegenerative and neuroinflammatory nature. Some, like Alzheimer's disease, Parkinson's disease, and multiple system atrophy, have been explored extensively, while others pertaining to metabolism not so much. However, emerging evidence points to hypothalamic inflammation mediated by microglia as a driver of metabolic dysregulations, particularly insulin resistance and type 2 diabetes mellitus. Here, we explore this connection further and examine pathways that underlie this relationship, including the IKKβ/NF-κβ, IRS-1/PI3K/Akt, mTOR-S6 Kinase, JAK/STAT, and PPAR-γ signaling pathways. We also investigate the role of non-coding RNAs, namely microRNAs and long non-coding RNAs, in insulin resistance related to neuroinflammation and their diagnostic and therapeutic potential. Finally, we explore therapeutics further, searching for both pharmacological and non-pharmacological interventions that can help mitigate microglial activation.
Collapse
Affiliation(s)
- Radwan Darwish
- School of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Yasmine Alcibahy
- School of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Shahd Bucheeri
- School of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Ashraf Albishtawi
- School of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Maya Tama
- School of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Jeevan Shetty
- Department of Biochemistry, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Alexandra E Butler
- School of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| |
Collapse
|
15
|
Aboutaleb AS, Allam A, Zaky HS, Harras MF, Farag FSAA, Abdel-Sattar SA, El-Said NT, Ahmed HI, Abd El-Mordy FM. Novel insights into the molecular mechanisms underlying anti-nociceptive effect of myricitrin against reserpine-induced fibromyalgia model in rats: Implication of SIRT1 and miRNAs. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118623. [PMID: 39059685 DOI: 10.1016/j.jep.2024.118623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/13/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Manilkara zapota (L.) P. Royen, also termed sapodilla or chikoo, is a significant plant in ethnomedicine because of its long history of traditional medical applications. In diverse cultures, sapodilla is believed to protect against oxidative stress, inflammation, and some chronic diseases because of its high antioxidant content. The naturally occurring antioxidant myricitrin (MYR) flavonoid is primarily found in the leaves and other plant parts of sapodilla and it is well-known for having therapeutic qualities and possible health advantages. AIM OF THE STUDY To appraise the possible impact of MYR on a rat model of reserpine-induced fibromyalgia (FM) and explore its mechanism of action. MATERIALS AND METHODS Isolation and identification of MYR with more than 99% purity from Manilkara zapota leaves were primarily done and confirmed through chromatographic and spectrophotometric techniques. To develop FM model, reserpine (RSP) was injected daily (1 mg/kg, s.c.) for three successive days. Then, MYR (10 mg/kg, i.p.) and pregabalin (PGB, 30 mg/kg, p.o.) were given daily for another five days. Behavioral changes were assessed through open field test (OFT), hot plate test, and forced swimming test (FST). Further analyses of different brain parameters and signaling pathways were performed to assess monoamines levels, oxidative stress, inflammatory response, apoptotic changes as well as silent information regulator 1 (SIRT1) and micro RNAs (miRNAs) expressions. RESULTS From High-Performance Liquid Chromatography (HPLC) analysis, the methanol extract of sapodilla leaves contains 166.17 μg/ml of MYR. Results of behavioral tests showed a significant improvement in RSP-induced nociceptive stimulation, reduced locomotion and exploration and depressive-like behavior by MYR. Biochemical analyses showed that MYR significantly ameliorated the RSP-induced imbalance in brain monoamine neurotransmitters. In addition, MYR significantly attenuated oxidative stress elicited by RSP via up-regulating nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) protein expressions, enhancing superoxide dismutase (SOD) and catalase (CAT) activities, and reducing malondialdehyde (MDA) content in brain. The RSP-provoked inflammatory response was also diminished by MYR treatment as shown by a significant decreased NOD-like receptor protein 3 (NLRP3) inflammasome expression along with reduced levels of interleukin 1 beta (IL-1β) and nuclear factor-κB (NF-κB). Furthermore, the anti-apoptotic activity of MYR was demonstrated by a marked rise in Bcl-2-associated X protein (BAX)/B cell lymphoma-2 (Bcl-2) ratio by lowering Bcl-2 while increasing BAX levels. In addition, MYR treatment significantly boosted the expression of SIRT1 deacetylase in RSP-treated animals. Interestingly, molecular docking showed the ability of MYR to form a stable complex in the binding site of SIRT1. Regarding miRNAs, MYR effectively ameliorated RSP-induced changes in miR-320 and miR-107 gene expressions. CONCLUSION Our findings afford new insights into the anti-nociceptive profile of MYR in the RSP-induced FM model in rats. The underlying mechanisms involved direct binding and activation of SIRT1 to influence different signaling cascades, including Nrf2 and NF-κB/NLRP3 together with modulation of miRNAs. However, more in-depth studies are needed before proposing MYR as a new clinically relevant drug in the management of FM.
Collapse
Affiliation(s)
- Amany S Aboutaleb
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Albatoul Allam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Heba S Zaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Marwa F Harras
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Fatma Sayed Abdel-Aal Farag
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Somaia A Abdel-Sattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt.
| | - Nermin T El-Said
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Hebatalla I Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Fatma Mohamed Abd El-Mordy
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
16
|
Carpi S, Daniele S, de Almeida JFM, Gabbia D. Recent Advances in miRNA-Based Therapy for MASLD/MASH and MASH-Associated HCC. Int J Mol Sci 2024; 25:12229. [PMID: 39596297 PMCID: PMC11595301 DOI: 10.3390/ijms252212229] [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: 10/03/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), is a growing health concern worldwide, affecting more than 1 billion adults. It may progress to metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and ultimately hepatocellular carcinoma (HCC). Emerging evidence has demonstrated the role in this transition of microRNAs (miRNAs), which regulate the expression of genes associated with lipid metabolism, inflammation, fibrosis, and cell proliferation. Specific miRNAs have been identified to exacerbate or mitigate fibrotic and carcinogenic processes in hepatic cells. The modulation of these miRNAs through synthetic mimics or inhibitors represents a promising therapeutic strategy. Preclinical models have demonstrated that miRNA-based therapies can attenuate liver inflammation, reduce fibrosis, and inhibit tumorigenesis, thus delaying or preventing the onset of HCC. However, challenges such as delivery mechanisms, off-target effects, and long-term safety remain to be addressed. This review, focusing on recently published preclinical and clinical studies, explores the pharmacological potential of miRNA-based interventions to prevent MASLD/MASH and progression toward HCC.
Collapse
Affiliation(s)
- Sara Carpi
- Department of Health Sciences, University ‘Magna Græcia’ of Catanzaro, 88100 Catanzaro, Italy
- NEST (National Enterprise for nanoScience and nanoTechnology), Istituto Nanoscienze-CNR and Scuola Normale Superiore, 41125 Modena, Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (S.D.); (J.F.M.d.A.)
| | | | - Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
17
|
Kumar A, Choudhary A, Munshi A. Epigenetic reprogramming of mtDNA and its etiology in mitochondrial diseases. J Physiol Biochem 2024; 80:727-741. [PMID: 38865050 DOI: 10.1007/s13105-024-01032-z] [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: 04/03/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024]
Abstract
Mitochondrial functionality and its regulation are tightly controlled through a balanced crosstalk between the nuclear and mitochondrial DNA interactions. Epigenetic signatures like methylation, hydroxymethylation and miRNAs have been reported in mitochondria. In addition, epigenetic signatures encoded by nuclear DNA are also imported to mitochondria and regulate the gene expression dynamics of the mitochondrial genome. Alteration in the interplay of these epigenetic modifications results in the pathogenesis of various disorders like neurodegenerative, cardiovascular, metabolic disorders, cancer, aging and senescence. These modifications result in higher ROS production, increased mitochondrial copy number and disruption in the replication process. In addition, various miRNAs are associated with regulating and expressing important mitochondrial gene families like COX, OXPHOS, ND and DNMT. Epigenetic changes are reversible and therefore therapeutic interventions like changing the target modifications can be utilized to repair or prevent mitochondrial insufficiency by reversing the changed gene expression. Identifying these mitochondrial-specific epigenetic signatures has the potential for early diagnosis and treatment responses for many diseases caused by mitochondrial dysfunction. In the present review, different mitoepigenetic modifications have been discussed in association with the development of various diseases by focusing on alteration in gene expression and dysregulation of specific signaling pathways. However, this area is still in its infancy and future research is warranted to draw better conclusions.
Collapse
Affiliation(s)
- Anil Kumar
- Department of Human Genetics and Molecular Medicines, Central University of Punjab, Bathinda, India
| | - Anita Choudhary
- Department of Human Genetics and Molecular Medicines, Central University of Punjab, Bathinda, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicines, Central University of Punjab, Bathinda, India.
| |
Collapse
|
18
|
Capetini VC, Quintanilha BJ, Garcia BREV, Rogero MM. Dietary modulation of microRNAs in insulin resistance and type 2 diabetes. J Nutr Biochem 2024; 133:109714. [PMID: 39097171 DOI: 10.1016/j.jnutbio.2024.109714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 07/13/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
The prevalence of type 2 diabetes is increasing worldwide. Various molecular mechanisms have been proposed to interfere with the insulin signaling pathway. Recent advances in proteomics and genomics indicate that one such mechanism involves the post-transcriptional regulation of insulin signaling by microRNA (miRNA). These noncoding RNAs typically induce messenger RNA (mRNA) degradation or translational repression by interacting with the 3' untranslated region (3'UTR) of target mRNA. Dietary components and patterns, which can either enhance or impair the insulin signaling pathway, have been found to regulate miRNA expression in both in vitro and in vivo studies. This review provides an overview of the current knowledge of how dietary components influence the expression of miRNAs related to the control of the insulin signaling pathway and discusses the potential application of these findings in precision nutrition.
Collapse
Affiliation(s)
- Vinícius Cooper Capetini
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil; Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, Institute of Pharmaceutical Science, Department of Pharmacology, King's College London, London, United Kingdom.
| | - Bruna Jardim Quintanilha
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil
| | - Bruna Ruschel Ewald Vega Garcia
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Marcelo Macedo Rogero
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil
| |
Collapse
|
19
|
Ataollahi Eshkoor S, Fanijavadi S. Dysbiosis-epigenetics-immune system interaction and ageing health problems. J Med Microbiol 2024; 73. [PMID: 39606883 DOI: 10.1099/jmm.0.001921] [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: 11/29/2024] Open
Abstract
Background. The growing interest in microbiota-epigenetics-immune system research stems from the understanding that microbiota, a group of micro-organisms colonized in the human body, can influence the gene expression through epigenetic mechanisms and interaction with the immune system. Epigenetics refers to changes in gene activity that are not caused by the alteration in the DNA sequence itself.Discussion. The clinical significance of this research lies in the potential to develop new therapies for diseases linked to the imbalance of these microbial species (dysbiosis), such as cancer and neurodegenerative diseases. The intricate interaction between microbiota and epigenetics involves the production of metabolites and signalling molecules that can impact our health by influencing immune responses, metabolism and inflammation. Understanding these interactions could lead to novel therapeutic strategies targeting microbiota-epigenetic pathways to improve health outcomes.Conclusion. In this context, we aim to review and emphasize the current knowledge and key concepts that link the microbiota to epigenetics and immune system function, exploring their relevance to the development and maintenance of homeostasis and susceptibility to different diseases later in life. We aim to elucidate key concepts concerning the interactions and potential effects among the human gut microbiota, epigenetics, the immune system and ageing diseases linked to dysbiosis.
Collapse
|
20
|
Yin QH, Hu JB, Zhou Q, Weng J, Shen ED, Wen F, Liu SL, Yin LL, Tong YJ, Long L, Tang KW, Bai ST, Ou LD. Unveiling miRNA30b's Role in Suppressing ADAM12 to Combat Triple-Negative Breast Cancer. Breast J 2024; 2024:5202941. [PMID: 39742357 PMCID: PMC11540880 DOI: 10.1155/2024/5202941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/25/2024] [Accepted: 10/15/2024] [Indexed: 01/03/2025]
Abstract
Background: Triple-negative breast cancer, a subtype of breast cancer, is characterized by a poor prognosis. Recent studies have shown that miRNA30b acts as an oncogene and is vital for the proliferation of malignancies across various systems. This study aimed to elucidate the impact of miRNA30b on the proliferation, migration, and invasion capabilities of breast cancer cells in vitro. Methods: Triple-negative breast cancer cell lines MDA-MB-231 were transiently transfected with miRNA30b inhibitor, mimic, or the negative control by Lipofectamine 2000. Successful transfection was confirmed by quantitative real-time polymerase chain reaction (qRT-PCR). Functional assays, including CCK8, clone formation, scratch, and transwell assays, were conducted to evaluate the proliferation, invasion, and migration ability of MDA-MB-231 cells in each group. The target protein of miRNA30b was determined using an online prediction data website, and the dual-luciferase assay confirmed whether there was a binding site between miRNA30b and ADAM12. The effect was further verified by Western blot analysis. Results: MDA-MB-231 cells were transfected with miRNA30b inhibitor, mimic, and negative control. miRNA30b expression was downregulated in the cells. Relative to the negative control group, miRNA30b expression significantly increased in the mimic group and decreased in the miRNA30b inhibitor group, with the differences being statistically significant. The miRNA30b mimic group exhibited a significant increase in miRNA30b expression and a corresponding promotion of cell proliferation, colony formation, and migration. Conversely, the miRNA30b inhibitor group displayed significantly reduced miRNA30b expression and suppressed cell proliferation, colony formation, and migration abilities compared to the negative control cells. Bioinformatics software predicted ADAM12 as a potential target of miRNA30b. Dual-luciferase assays confirmed the presence of a binding site between miRNA30b and ADAM12. Western blot analysis revealed that overexpression of miRNA30b downregulated ADAM12 expression in MDA-MB-231 cells. Conclusions: miRNA30b could promote proliferation, migration, and invasion of TNBC cell lines MDA-MB-231. The effect of miRNA30b on triple-negative breast cancer would be achieved partly at least through inhibiting the expression of ADAM12.
Collapse
Affiliation(s)
- Qing-hua Yin
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Jian-bing Hu
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Qiang Zhou
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Jie Weng
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Er-dong Shen
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Fang Wen
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Song-lian Liu
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Lei-lan Yin
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Ya-jun Tong
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Ling Long
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Ke-wei Tang
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Si-te Bai
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Lu-di Ou
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| |
Collapse
|
21
|
de Mello AS, Ferguson BS, Shebs-Maurine EL, Giotto FM. MicroRNA Biogenesis, Gene Regulation Mechanisms, and Availability in Foods. Noncoding RNA 2024; 10:52. [PMID: 39452838 PMCID: PMC11510440 DOI: 10.3390/ncrna10050052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNAs that control gene expression by degrading or repressing mRNA translation into proteins. Research recently suggested that food-derived miRNAs are bioavailable and may be absorbed in the gastrointestinal tract (GIT). Since these small RNAs may reach the circulation and organs, possible interactions with host genes will lead to epigenetic effects that alter metabolism. Therefore, from a precision nutrition standpoint, exogenous miRNAs may be essential in modulating health status. This review summarizes the process of miRNA biogenesis, the post-translational mechanisms of gene regulation, and their bioavailability in animal- and plant-derived foods.
Collapse
Affiliation(s)
- Amilton S. de Mello
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno 1664 N. Virginia St. Mail Stop 202, Reno, NV 89557, USA; (E.L.S.-M.); (F.M.G.)
| | - Bradley S. Ferguson
- Department of Nutrition, University of Nevada, 1664 N. Virginia St. Mail Stop 202, Reno, NV 89557, USA;
| | - Erica L. Shebs-Maurine
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno 1664 N. Virginia St. Mail Stop 202, Reno, NV 89557, USA; (E.L.S.-M.); (F.M.G.)
| | - Francine M. Giotto
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno 1664 N. Virginia St. Mail Stop 202, Reno, NV 89557, USA; (E.L.S.-M.); (F.M.G.)
- Department of Animal and Range Sciences, New Mexico State University, Knox Hall 220, MSC 3-I, Las Cruces, NM 88003, USA
| |
Collapse
|
22
|
Ha SE, Singh R, Jin B, Baek G, Jorgensen BG, Zogg H, Debnath S, Park HS, Cho H, Watkins CM, Cho S, Kim MS, Lee MY, Yu TY, Jeong JW, Ro S. miR-10a/b-5p-NCOR2 Regulates Insulin-Resistant Diabetes in Female Mice. Int J Mol Sci 2024; 25:10147. [PMID: 39337631 PMCID: PMC11432729 DOI: 10.3390/ijms251810147] [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: 08/24/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Gender and biological sex have distinct impacts on the pathogenesis of type 2 diabetes (T2D). Estrogen deficiency is known to predispose female mice to T2D. In our previous study, we found that a high-fat, high-sucrose diet (HFHSD) induces T2D in male mice through the miR-10b-5p/KLF11/KIT pathway, but not in females, highlighting hormonal disparities in T2D susceptibility. However, the underlying molecular mechanisms of this hormonal protection in females remain elusive. To address this knowledge gap, we utilized ovariectomized, estrogen-deficient female mice, fed them a HFHSD to induce T2D, and investigated the molecular mechanisms involved in estrogen-deficient diabetic female mice, relevant cell lines, and female T2D patients. Initially, female mice fed a HFHSD exhibited a delayed onset of T2D, but ovariectomy-induced estrogen deficiency promptly precipitated T2D without delay. Intriguingly, insulin (INS) was upregulated, while insulin receptor (INSR) and protein kinase B (AKT) were downregulated in these estrogen-deficient diabetic female mice, indicating insulin-resistant T2D. These dysregulations of INS, INSR, and AKT were mediated by a miR-10a/b-5p-NCOR2 axis. Treatment with miR-10a/b-5p effectively alleviated hyperglycemia in estrogen-deficient T2D female mice, while β-estradiol temporarily reduced hyperglycemia. Consistent with the murine findings, plasma samples from female T2D patients exhibited significant reductions in miR-10a/b-5p, estrogen, and INSR, but increased insulin levels. Our findings suggest that estrogen protects against insulin-resistant T2D in females through miR-10a/b-5p/NCOR2 pathway, indicating the potential therapeutic benefits of miR-10a/b-5p restoration in female T2D management.
Collapse
Affiliation(s)
- Se Eun Ha
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Rajan Singh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Byungchang Jin
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Gain Baek
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Brian G. Jorgensen
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Hannah Zogg
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Sushmita Debnath
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Hahn Sung Park
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Hayeong Cho
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Claudia Marie Watkins
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Sumin Cho
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Min-Seob Kim
- Department of Physiology, Wonkwang Digestive Disease Research Institute & Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (M.-S.K.); (M.Y.L.)
| | - Moon Young Lee
- Department of Physiology, Wonkwang Digestive Disease Research Institute & Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (M.-S.K.); (M.Y.L.)
| | - Tae Yang Yu
- Division of Endocrinology and Metabolism, Department of Medicine, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea; (T.Y.Y.); (J.W.J.)
| | - Jin Woo Jeong
- Division of Endocrinology and Metabolism, Department of Medicine, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea; (T.Y.Y.); (J.W.J.)
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
- RosVivo Therapeutics, Applied Research Facility, 1664 N. Virginia St., Reno, NV 89557, USA
| |
Collapse
|
23
|
Odimegwu CL, Uwaezuoke SN, Chikani UN, Mbanefo NR, Adiele KD, Nwolisa CE, Eneh CI, Ndiokwelu CO, Okpala SC, Ogbuka FN, Odo KE, Ohuche IO, Obiora-Izuka CE. Targeting the Epigenetic Marks in Type 2 Diabetes Mellitus: Will Epigenetic Therapy Be a Valuable Adjunct to Pharmacotherapy? Diabetes Metab Syndr Obes 2024; 17:3557-3576. [PMID: 39323929 PMCID: PMC11423826 DOI: 10.2147/dmso.s479077] [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] [Received: 05/20/2024] [Accepted: 08/03/2024] [Indexed: 09/27/2024] Open
Abstract
Although genetic, environmental, and lifestyle factors largely contribute to type 2 diabetes mellitus (T2DM) risk, the role of epigenetics in its pathogenesis is now well established. The epigenetic mechanisms in T2DM mainly consist of DNA methylation, histone modifications and regulation by noncoding RNAs (ncRNAs). For instance, DNA methylation at CpG islands in the promoter regions of specific genes encoding insulin signaling and glucose metabolism suppresses these genes. Modulating the enzyme mediators of these epigenetic marks aims to restore standard gene expression patterns and improve glycemic control. In targeting these epigenetic marks, using epigenetic drugs such as DNA methyltransferase (DNAMT), histone deacetylase (HDAC) and histone acetyltransferase (HAT) inhibitors has led to variable success in humans and experimental murine models. Specifically, the United States' Food and Drug Administration (US FDA) has approved DNAMT inhibitors like 5-azacytidine and 5-aza-2'-deoxycytidine for use in diabetic retinopathy: a T2DM microvascular complication. These DNAMT inhibitors block the genes for methylation of mitochondrial superoxide dismutase 2 (SOD2) and matrix metallopeptidase 9 (MMP-9): the epigenetic marks in diabetic retinopathy. Traditional pharmacotherapy with metformin also have epigenetic effects in T2DM and positively alter disease outcomes when combined with epigenetic drugs like DNAMT and HDAC inhibitors, raising the prospect of using epigenetic therapy as a valuable adjunct to pharmacotherapy. However, introducing small interfering RNAs (siRNAs) in cells to silence specific target genes remains in the exploratory phase. Future research should focus on regulating gene expression in T2DM using long noncoding RNA (lncRNA) molecules, another type of ncRNA. This review discusses the epigenetics of T2DM and that of its macro- and microvascular complications, and the potential benefits of combining epigenetic therapy with pharmacotherapy for optimal results.
Collapse
Affiliation(s)
- Chioma Laura Odimegwu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Samuel Nkachukwu Uwaezuoke
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ugo N Chikani
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ngozi Rita Mbanefo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ken Daberechi Adiele
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | - Chizoma Ihuarula Eneh
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Chibuzo Obiora Ndiokwelu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Somkenechi C Okpala
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Francis N Ogbuka
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Kenneth E Odo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | | |
Collapse
|
24
|
Rao G, Peng B, Zhang G, Fu X, Tian J, Tian Y. MicroRNAs in diabetic macroangiopathy. Cardiovasc Diabetol 2024; 23:344. [PMID: 39285459 PMCID: PMC11406791 DOI: 10.1186/s12933-024-02405-w] [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: 06/04/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Diabetic macroangiopathy is a leading cause of diabetes-related mortality worldwide. Both genetic and environmental factors, through a multitude of underlying molecular mechanisms, contribute to the pathogenesis of diabetic macroangiopathy. MicroRNAs (miRNAs), a class of non-coding RNAs known for their functional diversity and expression specificity, are increasingly recognized for their roles in the initiation and progression of diabetes and diabetic macroangiopathy. In this review, we will describe the biogenesis of miRNAs, and summarize their functions in diabetic macroangiopathy, including atherosclerosis, peripheral artery disease, coronary artery disease, and cerebrovascular disease, which are anticipated to provide new insights into future perspectives of miRNAs in basic, translational and clinical research, ultimately advancing the diagnosis, prevention, and treatment of diabetic macroangiopathy.
Collapse
Affiliation(s)
- Guocheng Rao
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, China
| | - Boqiang Peng
- Department of General Surgery and Gastric Cancer Center and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Guixiang Zhang
- Department of General Surgery and Gastric Cancer Center and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xianghui Fu
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, China.
| | - Jingyan Tian
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Tian
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
25
|
Ranhotra HS. Discrete interplay of gut microbiota L-tryptophan metabolites in host biology and disease. Mol Cell Biochem 2024; 479:2273-2290. [PMID: 37861881 DOI: 10.1007/s11010-023-04867-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/24/2023] [Indexed: 10/21/2023]
Abstract
The gut microbiota and the host maintain a conjoint relationship and together achieve optimal physiology via a multitude of interactive signalling cues. Dietary-derived L-tryptophan (L-trp) is enzymatically metabolized by the resident symbiotic gut microbiota to indole and various indole derivatives. Indole and indole metabolites secreted by the gut bacteria act locally in the intestinal cells as well as distally and modulate tissue-specific functions which are beneficial to the host. Functions attributed to these microbial indole metabolites in the host include regulation of intestinal permeability, immunity and mucosal roles, inflammation, and insulin sensitivity. On the other hand, dysregulation of gut microbiota L-trp metabolism compromises the optimal availability of indole and indole metabolites and can induce the onset of metabolic disorders, inflammation, liver steatosis, and decrease gut barrier integrity. Gut dysbiosis is regarded as one of the prime reasons for this deregulated microbial-derived indole metabolites. A number of indole metabolites from the gut bacteria have been identified recently displaying variable affinity towards xenobiotic nuclear receptors. Microbial metabolite mimicry concept can be used to design and develop novel indole-moiety-containing compounds with higher affinity towards the receptors and efficacy in preclinical studies. Such compounds may serve as therapeutic drugs in clinical trials in the future. In this article, I review L-trp metabolism in the host and gut microbiota and the various physiological functions, patho-physiologies associated with the microbial-released indole metabolites in the host, including the metabolite mimicry-based concept to develop tailored indole-containing novel experimental drugs.
Collapse
Affiliation(s)
- Harmit S Ranhotra
- Department of Biochemistry, St. Edmund's College, Shillong, 793 003, India.
| |
Collapse
|
26
|
He L, Wang X, Chen X. Unveiling the role of microRNAs in metabolic dysregulation of Gestational Diabetes Mellitus. Reprod Biol 2024; 24:100924. [PMID: 39013209 DOI: 10.1016/j.repbio.2024.100924] [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: 05/01/2024] [Revised: 05/26/2024] [Accepted: 06/05/2024] [Indexed: 07/18/2024]
Abstract
Gestational Diabetes Mellitus (GDM) presents a significant health concern globally, necessitating a comprehensive understanding of its metabolic intricacies for effective management. MicroRNAs (miRNAs) have emerged as pivotal regulators in GDM pathogenesis, influencing glucose metabolism, insulin signaling, and lipid homeostasis during pregnancy. Dysregulated miRNA expression, both upregulated and downregulated, contributes to GDM-associated metabolic abnormalities. Ethnic and temporal variations in miRNA expression underscore the multifaceted nature of GDM susceptibility. This review examines the dysregulation of miRNAs in GDM and their regulatory functions in metabolic disorders. We discuss the involvement of specific miRNAs in modulating key pathways implicated in GDM pathogenesis, such as glucose metabolism, insulin signaling, and lipid homeostasis. Furthermore, we explore the potential diagnostic and therapeutic implications of miRNAs in GDM management, highlighting the promise of miRNA-based interventions for mitigating the adverse consequences of GDM on maternal and offspring health.
Collapse
Affiliation(s)
- Ling He
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoli Wang
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyi Chen
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
27
|
Liu T, Zhu C, Duan Z, Ma P, Ma X, Fan D. Network Pharmacological Analysis Combined with Experimental Verification to Explore the Effect of Ginseng Polypeptide on the Improvement of Diabetes Symptoms in db/db Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18537-18551. [PMID: 39129180 DOI: 10.1021/acs.jafc.4c04949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Diabetes mellitus is a typical metabolic disease that has become a major threat to human health worldwide. Ginseng polypeptide (GP), a small molecule active substance isolated from ginseng, has shown positive hypoglycemic effects in preliminary studies. However, its mechanism in ameliorating multiorgan damage in db/db mice is unclear. In this study, we utilized network pharmacology, molecular docking, and animal experiments to explore the targets and biological mechanisms of GP to ameliorate multiorgan damage in T2DM. The results showed that GP improves T2DM by inhibiting inflammation and oxidative damage, thereby alleviating hyperglycemia, insulin resistance, and multiorgan damage in db/db mice. These effects are potentially mediated through the PI3K-Akt signaling pathway and the MAPK signaling pathway. This study establishes GP's efficacy in alleviating T2DM and provides a robust theoretical basis for the development of new drugs or functional foods for treating this disease.
Collapse
Affiliation(s)
- Tianzhu Liu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Pei Ma
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Xiaoxuan Ma
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| |
Collapse
|
28
|
Zhang L, Zhang L, Chen H, Xu X. The Interplay Between Cytokines and MicroRNAs to Regulate Metabolic Disorders. J Interferon Cytokine Res 2024; 44:337-348. [PMID: 39082185 DOI: 10.1089/jir.2024.0059] [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: 08/23/2024] Open
Abstract
Metabolic disorders represent significant public health challenges worldwide. Emerging evidence suggests that cytokines and microRNAs (miRNAs) play crucial roles in the pathogenesis of metabolic disorders by regulating various metabolic processes, including insulin sensitivity, lipid metabolism, and inflammation. This review provides a comprehensive overview of the intricate interplay between cytokines and miRNAs in the context of metabolic disorders, including obesity, type 2 diabetes, and cardiovascular diseases. We discuss how dysregulation of cytokine-miRNA networks contributes to the development and progression of metabolic disorders and explore the therapeutic potential of targeting these interactions for disease management.
Collapse
Affiliation(s)
- Li Zhang
- Department of Clinical Laboratory, The Second Staff Hospital of Wuhan Iron and Steel (Group) Corporation, Wuhan, China
| | - Li Zhang
- Department of Clinical Laboratory, The Second Staff Hospital of Wuhan Iron and Steel (Group) Corporation, Wuhan, China
| | - Huan Chen
- Department of Clinical Laboratory, Wuhan Institute of Technology Hospital, Wuhan Institute of Technology, Wuhan, China
| | - Xiangyong Xu
- Department of Clinical Laboratory, The Second Staff Hospital of Wuhan Iron and Steel (Group) Corporation, Wuhan, China
| |
Collapse
|
29
|
Sazdova I, Hadzi-Petrushev N, Keremidarska-Markova M, Stojchevski R, Sopi R, Shileiko S, Mitrokhin V, Gagov H, Avtanski D, Lubomirov LT, Mladenov M. SIRT-associated attenuation of cellular senescence in vascular wall. Mech Ageing Dev 2024; 220:111943. [PMID: 38762036 DOI: 10.1016/j.mad.2024.111943] [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: 04/13/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
This review focuses on the vital function that SIRT1 and other sirtuins play in promoting cellular senescence in vascular smooth muscle cells, which is a key element in the pathogenesis of vascular aging and associated cardiovascular diseases. Vascular aging is a gradual process caused by the accumulation of senescent cells, which results in increased vascular remodeling, stiffness, and diminished angiogenic ability. Such physiological alterations are characterized by a complex interplay of environmental and genetic variables, including oxidative stress and telomere attrition, which affect gene expression patterns and trigger cell growth arrest. SIRT1 has been highlighted for its potential to reduce cellular senescence through modulation of multiple signaling cascades, particularly the endothelial nitric oxide (eNOS)/NO signaling pathway. It also modulates cell cycle through p53 inactivation and suppresses NF-κB mediated expression of adhesive molecules at the vascular level. The study also examines the therapeutic potential of sirtuin modulation in vascular health, identifying SIRT1 and its sirtuin counterparts as potential targets for reducing vascular aging. This study sheds light on the molecular basis of vascular aging and the beneficial effects of sirtuins, paving the way for the development of tailored therapies aimed at enhancing vascular health and prolonging life.
Collapse
Affiliation(s)
- Iliyana Sazdova
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University 'St. Kliment Ohridski', Sofia 1504, Bulgaria
| | - Nikola Hadzi-Petrushev
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University, Skopje 1000, North Macedonia
| | - Milena Keremidarska-Markova
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University 'St. Kliment Ohridski', Sofia 1504, Bulgaria
| | - Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, 110 E 59th Street, New York, NY 10022, USA
| | - Ramadan Sopi
- Faculty of Medicine, University of Prishtina, Prishtina 10 000, Kosovo
| | - Stanislav Shileiko
- Department of Fundamental and Applied Physiology, Russian States Medical University, Moscow 117997, Russia
| | - Vadim Mitrokhin
- Department of Fundamental and Applied Physiology, Russian States Medical University, Moscow 117997, Russia
| | - Hristo Gagov
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University 'St. Kliment Ohridski', Sofia 1504, Bulgaria
| | - Dimitar Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, 110 E 59th Street, New York, NY 10022, USA
| | - Lubomir T Lubomirov
- Vascular Biology Research Group (RenEVA), Research Institute, Medical University-Varna, Varna, Bulgaria; Institute of Physiology and Pathophysiology, Faculty of Health - School of Medicine, Biomedical Center for Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany
| | - Mitko Mladenov
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University, Skopje 1000, North Macedonia; Department of Fundamental and Applied Physiology, Russian States Medical University, Moscow 117997, Russia.
| |
Collapse
|
30
|
Lino M, Garcia-Martin R, Muñoz VR, Ruiz GP, Nawaz A, Brandão BB, Dreyfus J, Pan H, Kahn CR. Multi-step regulation of microRNA expression and secretion into small extracellular vesicles by insulin. Cell Rep 2024; 43:114491. [PMID: 39002127 PMCID: PMC11363058 DOI: 10.1016/j.celrep.2024.114491] [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: 02/02/2024] [Revised: 06/05/2024] [Accepted: 06/26/2024] [Indexed: 07/15/2024] Open
Abstract
Tissues release microRNAs (miRNAs) in small extracellular vesicles (sEVs) including exosomes, which can regulate gene expression in distal cells, thus acting as modulators of local and systemic metabolism. Here, we show that insulin regulates miRNA secretion into sEVs from 3T3-L1 adipocytes and that this process is differentially regulated from cellular expression. Thus, of the 53 miRNAs upregulated and 66 miRNAs downregulated by insulin in 3T3-L1 sEVs, only 12 were regulated in parallel in cells. Insulin regulated this process in part by phosphorylating hnRNPA1, causing it to bind to AU-rich motifs in miRNAs, mediating their secretion into sEVs. Importantly, 43% of insulin-regulated sEV-miRNAs are implicated in obesity and insulin resistance. These include let-7 and miR-103, which we show regulate insulin signaling in AML12 hepatocytes. Together, these findings demonstrate an important layer to insulin's regulation of adipose biology and provide a mechanism of tissue crosstalk in obesity and other hyperinsulinemic states.
Collapse
Affiliation(s)
- Marsel Lino
- Joslin Diabetes Center, Harvard Medical School, Harvard University, Boston, MA, USA; Harvard Medical School, Harvard University, Boston, MA, USA
| | - Ruben Garcia-Martin
- Joslin Diabetes Center, Harvard Medical School, Harvard University, Boston, MA, USA; Harvard Medical School, Harvard University, Boston, MA, USA
| | - Vitor Rosetto Muñoz
- Joslin Diabetes Center, Harvard Medical School, Harvard University, Boston, MA, USA; Harvard Medical School, Harvard University, Boston, MA, USA
| | - Gabriel Palermo Ruiz
- Joslin Diabetes Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Allah Nawaz
- Joslin Diabetes Center, Harvard Medical School, Harvard University, Boston, MA, USA; Harvard Medical School, Harvard University, Boston, MA, USA
| | - Bruna Brasil Brandão
- Joslin Diabetes Center, Harvard Medical School, Harvard University, Boston, MA, USA; Harvard Medical School, Harvard University, Boston, MA, USA
| | - Jonathan Dreyfus
- Joslin Diabetes Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Hui Pan
- Joslin Diabetes Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - C Ronald Kahn
- Joslin Diabetes Center, Harvard Medical School, Harvard University, Boston, MA, USA; Harvard Medical School, Harvard University, Boston, MA, USA.
| |
Collapse
|
31
|
Krause C, Britsemmer JH, Bernecker M, Molenaar A, Taege N, Lopez-Alcantara N, Geißler C, Kaehler M, Iben K, Judycka A, Wagner J, Wolter S, Mann O, Pfluger P, Cascorbi I, Lehnert H, Stemmer K, Schriever SC, Kirchner H. Liver microRNA transcriptome reveals miR-182 as link between type 2 diabetes and fatty liver disease in obesity. eLife 2024; 12:RP92075. [PMID: 39037913 PMCID: PMC11262792 DOI: 10.7554/elife.92075] [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] [Indexed: 07/24/2024] Open
Abstract
Background The development of obesity-associated comorbidities such as type 2 diabetes (T2D) and hepatic steatosis has been linked to selected microRNAs in individual studies; however, an unbiased genome-wide approach to map T2D induced changes in the miRNAs landscape in human liver samples, and a subsequent robust identification and validation of target genes are still missing. Methods Liver biopsies from age- and gender-matched obese individuals with (n=20) or without (n=20) T2D were used for microRNA microarray analysis. The candidate microRNA and target genes were validated in 85 human liver samples, and subsequently mechanistically characterized in hepatic cells as well as by dietary interventions and hepatic overexpression in mice. Results Here, we present the human hepatic microRNA transcriptome of type 2 diabetes in liver biopsies and use a novel seed prediction tool to robustly identify microRNA target genes, which were then validated in a unique cohort of 85 human livers. Subsequent mouse studies identified a distinct signature of T2D-associated miRNAs, partly conserved in both species. Of those, human-murine miR-182-5 p was the most associated with whole-body glucose homeostasis and hepatic lipid metabolism. Its target gene LRP6 was consistently lower expressed in livers of obese T2D humans and mice as well as under conditions of miR-182-5 p overexpression. Weight loss in obese mice decreased hepatic miR-182-5 p and restored Lrp6 expression and other miR-182-5 p target genes. Hepatic overexpression of miR-182-5 p in mice rapidly decreased LRP6 protein levels and increased liver triglycerides and fasting insulin under obesogenic conditions after only seven days. Conclusions By mapping the hepatic miRNA-transcriptome of type 2 diabetic obese subjects, validating conserved miRNAs in diet-induced mice, and establishing a novel miRNA prediction tool, we provide a robust and unique resource that will pave the way for future studies in the field. As proof of concept, we revealed that the repression of LRP6 by miR-182-5 p, which promotes lipogenesis and impairs glucose homeostasis, provides a novel mechanistic link between T2D and non-alcoholic fatty liver disease, and demonstrate in vivo that miR-182-5 p can serve as a future drug target for the treatment of obesity-driven hepatic steatosis. Funding This work was supported by research funding from the Deutsche Forschungsgemeinschaft (KI 1887/2-1, KI 1887/2-2, KI 1887/3-1 and CRC-TR296), the European Research Council (ERC, CoG Yoyo LepReSens no. 101002247; PTP), the Helmholtz Association (Initiative and Networking Fund International Helmholtz Research School for Diabetes; MB) and the German Center for Diabetes Research (DZD Next Grant 82DZD09D1G).
Collapse
Affiliation(s)
- Christin Krause
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- German Center for Diabetes Research (DZD)MunichGermany
| | - Jan H Britsemmer
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- German Center for Diabetes Research (DZD)MunichGermany
| | - Miriam Bernecker
- German Center for Diabetes Research (DZD)MunichGermany
- Research Unit NeuroBiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz CentreMunichGermany
| | - Anna Molenaar
- German Center for Diabetes Research (DZD)MunichGermany
- Research Unit NeuroBiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz CentreMunichGermany
| | - Natalie Taege
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- German Center for Diabetes Research (DZD)MunichGermany
| | - Nuria Lopez-Alcantara
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- Institute for Experimental Endocrinology, University of LübeckLübeckGermany
| | - Cathleen Geißler
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
| | - Meike Kaehler
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus KielKielGermany
| | - Katharina Iben
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
| | - Anna Judycka
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
| | - Jonas Wagner
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Stefan Wolter
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Oliver Mann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Paul Pfluger
- German Center for Diabetes Research (DZD)MunichGermany
- Research Unit NeuroBiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz CentreMunichGermany
- Chair of Neurobiology of Diabetes, TUM School of Medicine, Technical University of MunichMunichGermany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus KielKielGermany
| | - Hendrik Lehnert
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- German Center for Diabetes Research (DZD)MunichGermany
- University Hospital of Coventry and WarwickshireCoventryUnited Kingdom
| | - Kerstin Stemmer
- German Center for Diabetes Research (DZD)MunichGermany
- Molecular Cell Biology, Institute of Theoretical Medicine, Faculty of Medicine, University of AugsburgAugsburgGermany
| | - Sonja C Schriever
- German Center for Diabetes Research (DZD)MunichGermany
- Research Unit NeuroBiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz CentreMunichGermany
| | - Henriette Kirchner
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- German Center for Diabetes Research (DZD)MunichGermany
| |
Collapse
|
32
|
Du J, Zhu Y, Yang X, Geng X, Xu Y, Zhang M, Zhang M. Berberine attenuates obesity-induced insulin resistance by inhibiting miR-27a secretion. Diabet Med 2024; 41:e15319. [PMID: 38711201 DOI: 10.1111/dme.15319] [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: 09/28/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 05/08/2024]
Abstract
INTRODUCTION Berberine (BBR) is an alkaloid found in plants. It has neuroprotective, anti-inflammatory and lipid-lowering activity. However, the efficacy of treatment with BBR and the mechanisms through which it acts need further study. AIMS This study investigated the therapeutic effects and the mechanism of action of BBR on obesity-induced insulin resistance in peripheral tissues. METHODS High-fat-fed C57BL/6J mice and low-fat-fed C57BL/6J mice with miR-27a overexpression were given BBR intervention (100 mg/kg, po), and the oral glucose tolerance test (OGTT) and insulin tolerance test (ITT) were performed. Palmitic acid-stimulated hypertrophic adipocyte models were treated with BBR (10 μM). Related indicators and protein expression levels were examined. RESULTS The AUCs of the OGTT and the ITT in the BBR intervention group were reduced significantly (p < 0.01) (p < 0.05), and the serum biochemical parameters, including FBG, TC, TG and LDL-C were significantly reduced after BBR intervention. In the in vitro experiments, the triglyceride level and volume of lipid droplets decreased significantly after BBR intervention (p < 0.01) (p < 0.05). Likewise, BBR ameliorates skeletal muscle and pancreas insulin signalling pathways in vivo and in vitro. DISCUSSION The results showed that BBR significantly ameliorated insulin resistance, reduced body weight and percent body fat and improved serum biochemical parameters in mice. Likewise, BBR reduced triglyceride level and lipid droplet volume in hypertrophic adipocytes, BBR improved obesity effectively. Meanwhile, BBR ameliorated the histomorphology of the pancreas, and skeletal muscle and pancreas insulin related signalling pathways of islets in in vitro and in vivo experiments. The results further demonstrated that BBR inhibited miR-27a levels in serum from obese mice and supernatant of hypertrophic adipocytes. miR-27a overexpression in low-fat fed mice indicated that miR-27a caused insulin resistance, and BBR intervention significantly improved the miR-27a induced insulin resistance status. CONCLUSION This study demonstrates the important role of BBR in obesity-induced peripheral insulin resistance and suggest that the mechanism of its effect may be inhibition of miR-27a secretion.
Collapse
Affiliation(s)
- Junda Du
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
- School of Pharmaceutical Science, Jilin University, Changchun, Jilin, China
| | - Yu Zhu
- Department of Ophthalmology of Jilin Province FAW General Hospital, Changchun, Jilin, China
| | - Xuehan Yang
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
| | - Xinru Geng
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
| | - Yang Xu
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
| | - Meishuang Zhang
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
| |
Collapse
|
33
|
Zacharjasz J, Sztachera M, Smuszkiewicz M, Piwecka M. Micromanaging the neuroendocrine system - A review on miR-7 and the other physiologically relevant miRNAs in the hypothalamic-pituitary axis. FEBS Lett 2024; 598:1557-1575. [PMID: 38858179 DOI: 10.1002/1873-3468.14948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/12/2024]
Abstract
The hypothalamic-pituitary axis is central to the functioning of the neuroendocrine system and essential for regulating physiological and behavioral homeostasis and coordinating fundamental body functions. The expanding line of evidence shows the indispensable role of the microRNA pathway in regulating the gene expression profile in the developing and adult hypothalamus and pituitary gland. Experiments provoking a depletion of miRNA maturation in the context of the hypothalamic-pituitary axis brought into focus a prominent involvement of miRNAs in neuroendocrine functions. There are also a few individual miRNAs and miRNA families that have been studied in depth revealing their crucial role in mediating the regulation of fundamental processes such as temporal precision of puberty timing, hormone production, fertility and reproduction capacity, and energy balance. Among these miRNAs, miR-7 was shown to be hypothalamus-enriched and the top one highly expressed in the pituitary gland, where it has a profound impact on gene expression regulation. Here, we review miRNA profiles, knockout phenotypes, and miRNA interaction (targets) in the hypothalamic-pituitary axis that advance our understanding of the roles of miRNAs in mammalian neurosecretion and related physiology.
Collapse
Affiliation(s)
- Julian Zacharjasz
- Department of Non-coding RNAs, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznań, Poland
| | - Marta Sztachera
- Department of Non-coding RNAs, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznań, Poland
| | - Michał Smuszkiewicz
- Department of Non-coding RNAs, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznań, Poland
| | - Monika Piwecka
- Department of Non-coding RNAs, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznań, Poland
| |
Collapse
|
34
|
Cao X, Jiang Z, Bu X, Li Q, Tian Y, Xu Z, Zhang B, Yuan X. MicroRNA-204-5p Attenuates Oxidative Stress, Apoptosis and Inflammation by Targeting TXNIP in Diabetic Cataract. Biochem Genet 2024:10.1007/s10528-024-10863-w. [PMID: 38896379 DOI: 10.1007/s10528-024-10863-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Diabetic cataract (DC) is a major cause of blindness in diabetic patients and it is characterized by early onset and rapid progression. MiR-204-5p was previously identified as one of the top five down-regulated miRNAs in human DC lens tissues. We aimed to determine the expression of miR-204-5p in human lens epithelial cells (HLECs) and explore its effects and mechanisms in regulating the progression of DC. The expression of miR-204-5p in the anterior capsules of DC patients and HLECs was examined by RT-qPCR. Bioinformatics tools were then used to identify the potential target of miR-204-5p. The relationship between miR-204-5p and the target gene was confirmed through a dual luciferase reporter assay. Additionally, the regulatory mechanism of oxidative stress, apoptosis, and inflammation in DC was investigated by overexpressing miR-204-5p using miR-204-5p agomir. The expression of miR-204-5p was downregulated in the anterior capsules of DC patients and HLECs. Overexpression of miR-204-5p reduced ROS levels, pro-apoptosis genes (Bid, Bax, caspase-3), and IL-1β production in HG-treated HLECs. TXNIP was the direct target of miR-204-5p by dual luciferase reporter assay. Therefore, this study demonstrated that miR-204-5p effectively reduced oxidative damage, apoptosis, and inflammation in HLECs under HG conditions by targeting TXNIP. Targeting miR-204-5p could be a promising therapeutic strategy for the potential treatment of DC.
Collapse
Affiliation(s)
- Xiang Cao
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, 300020, China
- Department of Ophthalmology, Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, 212002, China
| | - Zhixin Jiang
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, 300020, China
| | - Xiaofei Bu
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, 300020, China
| | - Qingyu Li
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, 300020, China
| | - Ye Tian
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, 300020, China
| | - Zijiao Xu
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, 300020, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Boyang Zhang
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, 300020, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiaoyong Yuan
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, 300020, China.
| |
Collapse
|
35
|
Li S, Xiong F, Zhang S, Liu J, Gao G, Xie J, Wang Y. Oligonucleotide therapies for nonalcoholic steatohepatitis. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102184. [PMID: 38665220 PMCID: PMC11044058 DOI: 10.1016/j.omtn.2024.102184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Nonalcoholic steatohepatitis (NASH) represents a severe disease subtype of nonalcoholic fatty liver disease (NAFLD) that is thought to be highly associated with systemic metabolic abnormalities. It is characterized by a series of substantial liver damage, including hepatocellular steatosis, inflammation, and fibrosis. The end stage of NASH, in some cases, may result in cirrhosis and hepatocellular carcinoma (HCC). Nowadays a large number of investigations are actively under way to test various therapeutic strategies, including emerging oligonucleotide drugs (e.g., antisense oligonucleotide, small interfering RNA, microRNA, mimic/inhibitor RNA, and small activating RNA) that have shown high potential in treating this fatal liver disease. This article systematically reviews the pathogenesis of NASH/NAFLD, the promising druggable targets proven by current studies in chemical compounds or biological drug development, and the feasibility and limitations of oligonucleotide-based therapeutic approaches under clinical or pre-clinical studies.
Collapse
Affiliation(s)
- Sixu Li
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610066, China
| | - Feng Xiong
- Department of Cardiology, The Third People’s Hospital of Chengdu, Chengdu 610031, China
| | - Songbo Zhang
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Jinghua Liu
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Viral Vector Core, University of Massachusetts Chan Medical, School, Worcester, MA 01605, USA
| | - Jun Xie
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Viral Vector Core, University of Massachusetts Chan Medical, School, Worcester, MA 01605, USA
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610066, China
| |
Collapse
|
36
|
Patra D, Ramprasad P, Sharma S, Dey U, Kumar V, Singh S, Dasgupta S, Kumar A, Tikoo K, Pal D. Adipose tissue macrophage-derived microRNA-210-3p disrupts systemic insulin sensitivity by silencing GLUT4 in obesity. J Biol Chem 2024; 300:107328. [PMID: 38679332 PMCID: PMC11145551 DOI: 10.1016/j.jbc.2024.107328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024] Open
Abstract
Management of chronic obesity-associated metabolic disorders is a key challenge for biomedical researchers. During chronic obesity, visceral adipose tissue (VAT) undergoes substantial transformation characterized by a unique lipid-rich hypoxic AT microenvironment which plays a crucial role in VAT dysfunction, leading to insulin resistance (IR) and type 2 diabetes. Here, we demonstrate that obese AT microenvironment triggers the release of miR-210-3p microRNA-loaded extracellular vesicles from adipose tissue macrophages, which disseminate miR-210-3p to neighboring adipocytes, skeletal muscle cells, and hepatocytes through paracrine and endocrine actions, thereby influencing insulin sensitivity. Moreover, EVs collected from Dicer-silenced miR-210-3p-overexpressed bone marrow-derived macrophages induce glucose intolerance and IR in lean mice. Mechanistically, miR-210-3p interacts with the 3'-UTR of GLUT4 mRNA and silences its expression, compromising cellular glucose uptake and insulin sensitivity. Therapeutic inhibition of miR-210-3p in VAT notably rescues high-fat diet-fed mice from obesity-induced systemic glucose intolerance. Thus, targeting adipose tissue macrophage-specific miR-210-3p during obesity could be a promising strategy for managing IR and type 2 diabetes.
Collapse
Affiliation(s)
- Debarun Patra
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Palla Ramprasad
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Shivam Sharma
- Department of Pharmacology and Toxicology, NIPER, S.A.S. Nagar, Punjab, India
| | - Upalabdha Dey
- Department of Molecular Biology & Biotechnology, Tezpur University, Tezpur, Assam, India
| | - Vinod Kumar
- Department of Pharmacology and Toxicology, NIPER, S.A.S. Nagar, Punjab, India
| | - Satpal Singh
- Department of Gastro Surgery, DMC&H, Ludhiana, Punjab, India
| | - Suman Dasgupta
- Department of Molecular Biology & Biotechnology, Tezpur University, Tezpur, Assam, India
| | - Aditya Kumar
- Department of Molecular Biology & Biotechnology, Tezpur University, Tezpur, Assam, India
| | - Kulbhushan Tikoo
- Department of Pharmacology and Toxicology, NIPER, S.A.S. Nagar, Punjab, India
| | - Durba Pal
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India.
| |
Collapse
|
37
|
Bonet ML, Ribot J, Sánchez J, Palou A, Picó C. Early Life Programming of Adipose Tissue Remodeling and Browning Capacity by Micronutrients and Bioactive Compounds as a Potential Anti-Obesity Strategy. Cells 2024; 13:870. [PMID: 38786092 PMCID: PMC11120104 DOI: 10.3390/cells13100870] [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: 04/09/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
The early stages of life, especially the period from conception to two years, are crucial for shaping metabolic health and the risk of obesity in adulthood. Adipose tissue (AT) plays a crucial role in regulating energy homeostasis and metabolism, and brown AT (BAT) and the browning of white AT (WAT) are promising targets for combating weight gain. Nutritional factors during prenatal and early postnatal stages can influence the development of AT, affecting the likelihood of obesity later on. This narrative review focuses on the nutritional programming of AT features. Research conducted across various animal models with diverse interventions has provided insights into the effects of specific compounds on AT development and function, influencing the development of crucial structures and neuroendocrine circuits responsible for energy balance. The hormone leptin has been identified as an essential nutrient during lactation for healthy metabolic programming against obesity development in adults. Studies have also highlighted that maternal supplementation with polyunsaturated fatty acids (PUFAs), vitamin A, nicotinamide riboside, and polyphenols during pregnancy and lactation, as well as offspring supplementation with myo-inositol, vitamin A, nicotinamide riboside, and resveratrol during the suckling period, can impact AT features and long-term health outcomes and help understand predisposition to obesity later in life.
Collapse
Affiliation(s)
- M. Luisa Bonet
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), University of the Balearic Islands, 07122 Palma, Spain
| | - Joan Ribot
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Juana Sánchez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), University of the Balearic Islands, 07122 Palma, Spain
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), University of the Balearic Islands, 07122 Palma, Spain
| |
Collapse
|
38
|
Zhu L, Jiao H, Gao W, Gong P, Shi C, Zhang F, Zhao J, Lu X, Liu B, Luo J. MiR-103-5p deficiency suppresses lipid accumulation via upregulating PLSCR4 and its host gene PANK3 in goat mammary epithelial cells. Int J Biol Macromol 2024; 267:131240. [PMID: 38583827 DOI: 10.1016/j.ijbiomac.2024.131240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/09/2024] [Accepted: 03/27/2024] [Indexed: 04/09/2024]
Abstract
Lipids are intimately related to the unique flavor and nutritional values of goat milk. MicroRNAs (miRNA) participate in the regulation of various biological functions, including the synthesis and degradation of lipids. Several studies have shown that miR-103 is involved in the regulation of lipid metabolism, however, the molecular mechanism by which miR-103 regulates lipid metabolism in goat mammary gland is poorly understood. In this study, miR-103 was knocked out in goat mammary epithelial cells (GMECs) by CRISPR/Cas9, and the accumulation of lipid droplets, triglycerides, and cholesterol in the cells was suppressed subsequently. Overexpression or knockdown of miR-103-5p and miR-103-3p in GMECs revealed that it was miR-103-5p that promoted lipid accumulation but not miR-103-3p. In addition, Pantothenate Kinase 3 (PANK3), the host gene of miR-103, and Phospholipid Scramblase 4 (PLSCR4) were identified as the target genes of miR-103-5p by dual fluorescein and miRNA pulldown. Furthermore, we identified that cellular lipid levels were negatively regulated by PANK3 and PLSCR4. Lastly, in miR-103 knockout GMECs, the knockdown of PANK and PLSCR4 rescued the lipid accumulation. These findings suggest that miR-103-5p promotes lipid accumulation by targeting PLSCR4 and the host gene PANK3 in GMECs, providing new insights for the regulation of goat milk lipids via miRNAs.
Collapse
Affiliation(s)
- Lu Zhu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Hongyun Jiao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Wenchang Gao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Ping Gong
- Institute of Animal Husbandry Quality Standards, Xinjiang Academy of Animal Sciences, China
| | - Chenbo Shi
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Fuhong Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Jianqing Zhao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xuefeng Lu
- Institute of Animal Husbandry Quality Standards, Xinjiang Academy of Animal Sciences, China
| | - Baolong Liu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Jun Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
39
|
Missong H, Joshi R, Khullar N, Thareja S, Navik U, Bhatti GK, Bhatti JS. Nutrient-epigenome interactions: Implications for personalized nutrition against aging-associated diseases. J Nutr Biochem 2024; 127:109592. [PMID: 38325612 DOI: 10.1016/j.jnutbio.2024.109592] [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: 10/15/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
Aging is a multifaceted process involving genetic and environmental interactions often resulting in epigenetic changes, potentially leading to aging-related diseases. Various strategies, like dietary interventions and calorie restrictions, have been employed to modify these epigenetic landscapes. A burgeoning field of interest focuses on the role of microbiota in human health, emphasizing system biology and computational approaches. These methods help decipher the intricate interplay between diet and gut microbiota, facilitating the creation of personalized nutrition strategies. In this review, we analysed the mechanisms related to nutritional interventions while highlighting the influence of dietary strategies, like calorie restriction and intermittent fasting, on microbial composition and function. We explore how gut microbiota affects the efficacy of interventions using tools like multi-omics data integration, network analysis, and machine learning. These tools enable us to pinpoint critical regulatory elements and generate individualized models for dietary responses. Lastly, we emphasize the need for a deeper comprehension of nutrient-epigenome interactions and the potential of personalized nutrition informed by individual genetic and epigenetic profiles. As knowledge and technology advance, dietary epigenetics stands on the cusp of reshaping our strategy against aging and related diseases.
Collapse
Affiliation(s)
- Hemi Missong
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Riya Joshi
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
40
|
Mitra T, Gulati R, Ramachandran K, Rajiv R, Enninga EAL, Pierret CK, Kumari R S, Janardhanan R. Endocrine disrupting chemicals: gestational diabetes and beyond. Diabetol Metab Syndr 2024; 16:95. [PMID: 38664841 PMCID: PMC11046910 DOI: 10.1186/s13098-024-01317-9] [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] [Received: 10/15/2023] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Gestational Diabetes Mellitus (GDM) has been on the rise for the last two decades along with the growing incidence of obesity. The ubiquitous use of Endocrine-Disrupting Chemicals (EDCs) worldwide has been associated with this increase in GDM incidence. Epigenetic modifications such as DNA methylation, histone acetylation, and methylation have been associated with prenatal exposure to EDCs. EDC exposure can also drive a sustained disruption of the hypothalamus-pituitary-thyroid axis and various other signaling pathways such as thyroid signaling, PPARγ signaling, PI3K-AKT signaling. This disruption leads to impaired glucose metabolism, insulin resistance as well as β-cell dysfunction, which culminate into GDM. Persistent EDC exposure in pregnant women also increases adipogenesis, which results in gestational weight gain. Importantly, pregnant mothers transfer these EDCs to the fetus via the placenta, thus leading to other pregnancy-associated complications such as intrauterine growth restriction (IUGR), and large for gestational age neonates. Furthermore, this early EDC exposure of the fetus increases the susceptibility of the infant to metabolic diseases in early life. The transgenerational impact of EDCs is also associated with higher vascular tone, cognitive aberrations, and enhanced susceptibility to lifestyle disorders including reproductive health anomalies. The review focuses on the impact of environmental toxins in inducing epigenetic alterations and increasing the susceptibility to metabolic diseases during pregnancy needs to be extensively studied such that interventions can be developed to break this vicious cycle. Furthermore, the use of EDC-associated ExomiRs from the serum of patients can help in the early diagnosis of GDM, thereby leading to triaging of patients based on increasing risk factor of the clinicopathological condition.
Collapse
Affiliation(s)
- Tridip Mitra
- Division of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, 603 203, Kattankulathur, Tamil Nadu, India
| | - Richa Gulati
- Division of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, 603 203, Kattankulathur, Tamil Nadu, India
| | - Krithika Ramachandran
- Division of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, 603 203, Kattankulathur, Tamil Nadu, India
| | - Rohan Rajiv
- Dietrich School of Arts and Sciences, University of Pittsburgh, 15260, Pittsburgh, PA, USA
| | | | - Chris K Pierret
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Sajeetha Kumari R
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, 603 203, Kattankulathur, Tamil Nadu, India
| | - Rajiv Janardhanan
- Division of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, 603 203, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
41
|
McCarty KJ, DeCarlo AN, Ricks RE, Pratt SL, Long NM. Effects of maternal nutrient restriction during gestation on bovine serum microRNA abundance. Anim Reprod Sci 2024; 263:107435. [PMID: 38401394 DOI: 10.1016/j.anireprosci.2024.107435] [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: 05/11/2023] [Revised: 02/05/2024] [Accepted: 02/11/2024] [Indexed: 02/26/2024]
Abstract
The objective was to determine the effects of maternal nutrient restriction during gestation on serum microRNA (miRNA) abundance in cattle. Primiparous Angus-cross cows (n=22) were fed either control (CON; to gain 1 Kg/week) or nutrient restricted (NR; 0.55% NEm) diets based on National Research Council requirements. On day 30 of gestation, cows were blocked by body condition and randomly assigned to one of three diets: CON (n=8) days 30-190; NR (n=7) days 30-110 followed by CON days 110-190 (NR/C); or CON (n=7) days 30-110 followed by NR days 110-190 (C/NR). At 190 days of gestation, maternal serum was collected for RNA isolation and analyzed using a miRNA microarray of known Bos taurus sequences. Data were normalized using LOWESS and analyzed via ANOVA. At 190 days of gestation, 16 miRNAs exhibited differential abundance (P<0.05) between treatments. Cows that underwent NR, irrespective of when the insult occurred, had downregulated bta-miR-126-3p compared to CON cows. Bta-miR-16b was downregulated and three miRNAs upregulated in NR/C compared to C/NR and CON cows. Additionally, seven miRNAs were downregulated and four miRNAs upregulated in C/NR compared to NR/C and CON cows. Comparison of NR/C and C/NR cows revealed three differentially abundant (P<0.04) miRNAs (bta-miR-2487_L-2R-3_1ss15CT, bta-miR-215, and bta-miR-760-5p). Top KEGG pathway enrichment of target genes included: pathways in cancer, PI3K-Akt signaling, focal adhesion, Ras signaling, proteoglycans in cancer, and MAPK signaling. In summary, maternal nutrient restriction altered serum miRNA abundance profiles irrespective of the time at which the nutritional insult was induced.
Collapse
Affiliation(s)
- K J McCarty
- Department of Animal and Veterinary Sciences, Clemson University, Clemson, SC 29634, USA
| | - A N DeCarlo
- Department of Animal and Veterinary Sciences, Clemson University, Clemson, SC 29634, USA
| | - R E Ricks
- Department of Animal and Veterinary Sciences, Clemson University, Clemson, SC 29634, USA
| | - S L Pratt
- Department of Animal and Veterinary Sciences, Clemson University, Clemson, SC 29634, USA
| | - N M Long
- Department of Animal and Veterinary Sciences, Clemson University, Clemson, SC 29634, USA.
| |
Collapse
|
42
|
Liu J, Meng L, Liu Z, Lu M, Wang R. Identification of HDAC9 and ARRDC4 as potential biomarkers and targets for treatment of type 2 diabetes. Sci Rep 2024; 14:7083. [PMID: 38528189 PMCID: PMC10963792 DOI: 10.1038/s41598-024-57794-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 03/21/2024] [Indexed: 03/27/2024] Open
Abstract
We aimed to identify the key potential insulin resistance (IR)-related genes and investigate their correlation with immune cell infiltration in type 2 diabetes (T2D). The GSE78721 dataset (68 diabetic patients and 62 controls) was downloaded from the Gene Expression Omnibus database and utilized for single-sample gene set enrichment analysis. IR-related genes were obtained from the Comparative Toxicology Genetics Database, and the final IR-differentially expressed genes (DEGs) were screened by intersecting with the DEGs obtained from the GSE78721 datasets. Functional enrichment analysis was performed, and the networks of the target gene with microRNA, transcription factor, and drug were constructed. Hub genes were identified based on a protein-protein interaction network. Least absolute shrinkage and selection operator regression and Random Forest and Boruta analysis were combined to screen diagnostic biomarkers in T2D, which were validated using the GSE76894 (19 diabetic patients and 84 controls) and GSE9006 (12 diabetic patients and 24 controls) datasets. Quantitative real-time polymerase chain reaction was performed to validate the biomarker expression in IR mice and control mice. In addition, infiltration of immune cells in T2D and their correlation with the identified markers were computed using CIBERSORT. We identified differential immune gene set regulatory T-cells in the GSE78721 dataset, and T2D samples were assigned into three clusters based on immune infiltration. A total of 2094 IR-DEGs were primarily enriched in response to endoplasmic reticulum stress. Importantly, HDAC9 and ARRDC4 were identified as markers of T2D and associated with different levels of immune cell infiltration. HDAC9 mRNA level were higher in the IR mice than in control mice, while ARRDC4 showed the opposite trend. In summary, we discovered potential vital biomarkers that contribute to immune cell infiltration associated with IR, which offers a new sight of immunotherapy for T2D.
Collapse
Affiliation(s)
- Jing Liu
- Endocrinology Department, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Lingzhen Meng
- General Medical Department, The Fourth Hospital of Hebei Medical University, No.12 Jiankang Road, Shijiazhuang, 050000, People's Republic of China
| | - Zhihong Liu
- Endocrinology Department, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, People's Republic of China.
| | - Ming Lu
- Medical Department, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Ruiying Wang
- Endocrinology Department, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, People's Republic of China
| |
Collapse
|
43
|
La Sala L, Carlini V, Conte C, Macas-Granizo MB, Afzalpour E, Martin-Delgado J, D'Anzeo M, Pedretti RFE, Naselli A, Pontiroli AE, Cappato R. Metabolic disorders affecting the liver and heart: Therapeutic efficacy of miRNA-based therapies? Pharmacol Res 2024; 201:107083. [PMID: 38309383 DOI: 10.1016/j.phrs.2024.107083] [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: 10/14/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
Liver and heart disease are major causes of death worldwide. It is known that metabolic alteration causing type 2 diabetes (T2D) and Nonalcoholic fatty liver (NAFLD) coupled with a derangement in lipid homeostasis, may exacerbate hepatic and cardiovascular diseases. Some pharmacological treatments can mitigate organ dysfunctions but the important side effects limit their efficacy leading often to deterioration of the tissues. It needs to develop new personalized treatment approaches and recent progresses of engineered RNA molecules are becoming increasingly viable as alternative treatments. This review outlines the current use of antisense oligonucleotides (ASOs), RNA interference (RNAi) and RNA genome editing as treatment for rare metabolic disorders. However, the potential for small non-coding RNAs to serve as therapeutic agents for liver and heart diseases is yet to be fully explored. Although miRNAs are recognized as biomarkers for many diseases, they are also capable of serving as drugs for medical intervention; several clinical trials are testing miRNAs as therapeutics for type 2 diabetes, nonalcoholic fatty liver as well as cardiac diseases. Recent advances in RNA-based therapeutics may potentially facilitate a novel application of miRNAs as agents and as druggable targets. In this work, we sought to summarize the advancement and advantages of miRNA selective therapy when compared to conventional drugs. In particular, we sought to emphasise druggable miRNAs, over ASOs or other RNA therapeutics or conventional drugs. Finally, we sought to address research questions related to efficacy, side-effects, and range of use of RNA therapeutics. Additionally, we covered hurdles and examined recent advances in the use of miRNA-based RNA therapy in metabolic disorders such as diabetes, liver, and heart diseases.
Collapse
Affiliation(s)
- Lucia La Sala
- IRCCS MultiMedica, 20138 Milan, Italy; Dept. of Biomedical Sciences for Health, University of Milan, Milan, Italy.
| | | | - Caterina Conte
- IRCCS MultiMedica, 20138 Milan, Italy; Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| | | | - Elham Afzalpour
- Dept. of Biomedical Sciences and Clinic, University of Milan, Milan, Italy
| | - Jimmy Martin-Delgado
- Hospital Luis Vernaza, Junta de Beneficiencia de Guayaquil, 090603 Guayaquil, Ecuador; Instituto de Investigacion e Innovacion en Salud Integral, Universidad Catolica de Santiago de Guayaquil, Guayaquil 090603, Ecuador
| | - Marco D'Anzeo
- AUO delle Marche, SOD Medicina di Laboratorio, Ancona, Italy
| | | | | | | | | |
Collapse
|
44
|
Dong H, Sun Y, Nie L, Cui A, Zhao P, Leung WK, Wang Q. Metabolic memory: mechanisms and diseases. Signal Transduct Target Ther 2024; 9:38. [PMID: 38413567 PMCID: PMC10899265 DOI: 10.1038/s41392-024-01755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/29/2024] Open
Abstract
Metabolic diseases and their complications impose health and economic burdens worldwide. Evidence from past experimental studies and clinical trials suggests our body may have the ability to remember the past metabolic environment, such as hyperglycemia or hyperlipidemia, thus leading to chronic inflammatory disorders and other diseases even after the elimination of these metabolic environments. The long-term effects of that aberrant metabolism on the body have been summarized as metabolic memory and are found to assume a crucial role in states of health and disease. Multiple molecular mechanisms collectively participate in metabolic memory management, resulting in different cellular alterations as well as tissue and organ dysfunctions, culminating in disease progression and even affecting offspring. The elucidation and expansion of the concept of metabolic memory provides more comprehensive insight into pathogenic mechanisms underlying metabolic diseases and complications and promises to be a new target in disease detection and management. Here, we retrace the history of relevant research on metabolic memory and summarize its salient characteristics. We provide a detailed discussion of the mechanisms by which metabolic memory may be involved in disease development at molecular, cellular, and organ levels, with emphasis on the impact of epigenetic modulations. Finally, we present some of the pivotal findings arguing in favor of targeting metabolic memory to develop therapeutic strategies for metabolic diseases and provide the latest reflections on the consequences of metabolic memory as well as their implications for human health and diseases.
Collapse
Affiliation(s)
- Hao Dong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuezhang Sun
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lulingxiao Nie
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Aimin Cui
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Pengfei Zhao
- Periodontology and Implant Dentistry Division, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Wai Keung Leung
- Periodontology and Implant Dentistry Division, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Qi Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
45
|
Luo Y, Li C. Advances in Research Related to MicroRNA for Diabetic Retinopathy. J Diabetes Res 2024; 2024:8520489. [PMID: 38375094 PMCID: PMC10876316 DOI: 10.1155/2024/8520489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/21/2023] [Accepted: 01/27/2024] [Indexed: 02/21/2024] Open
Abstract
Diabetic retinopathy (DR) is a severe microvascular complication of diabetes and is one of the primary causes of blindness in the working-age population in Europe and the United States. At present, no cure is available for DR, but early detection and timely intervention can prevent the rapid progression of the disease. Several treatments for DR are known, primarily ophthalmic treatment based on glycemia, blood pressure, and lipid control, which includes laser photocoagulation, glucocorticoids, vitrectomy, and antivascular endothelial growth factor (anti-VEGF) medications. Despite the clinical efficacy of the aforementioned therapies, none of them can entirely shorten the clinical course of DR or reverse retinopathy. MicroRNAs (miRNAs) are vital regulators of gene expression and participate in cell growth, differentiation, development, and apoptosis. MicroRNAs have been shown to play a significant role in DR, particularly in the molecular mechanisms of inflammation, oxidative stress, and neurodegeneration. The aim of this review is to systematically summarize the signaling pathways and molecular mechanisms of miRNAs involved in the occurrence and development of DR, mainly from the pathogenesis of oxidative stress, inflammation, and neovascularization. Meanwhile, this article also discusses the research progress and application of miRNA-specific therapies for DR.
Collapse
Affiliation(s)
- Yahan Luo
- Shanghai TCM-Integrated Hospital, Shanghai University of TCM, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunxia Li
- Shanghai TCM-Integrated Hospital, Shanghai University of TCM, Shanghai, China
| |
Collapse
|
46
|
Zailaie SA, Khoja BB, Siddiqui JJ, Mawardi MH, Heaphy E, Aljagthmi A, Sergi CM. Investigating the Role of Non-Coding RNA in Non-Alcoholic Fatty Liver Disease. Noncoding RNA 2024; 10:10. [PMID: 38392965 PMCID: PMC10891858 DOI: 10.3390/ncrna10010010] [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: 12/12/2023] [Revised: 01/22/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Non-coding RNAs (ncRNAs) are RNA molecules that do not code for protein but play key roles in regulating cellular processes. NcRNAs globally affect gene expression in diverse physiological and pathological contexts. Functionally important ncRNAs act in chromatin modifications, in mRNA stabilization and translation, and in regulation of various signaling pathways. Non-alcoholic fatty liver disease (NAFLD) is a set of conditions caused by the accumulation of triacylglycerol in the liver. Studies of ncRNA in NAFLD are limited but have demonstrated that ncRNAs play a critical role in the pathogenesis of NAFLD. In this review, we summarize NAFLD's pathogenesis and clinical features, discuss current treatment options, and review the involvement of ncRNAs as regulatory molecules in NAFLD and its progression to non-alcoholic steatohepatitis (NASH). In addition, we highlight signaling pathways dysregulated in NAFLD and review their crosstalk with ncRNAs. Having a thorough understanding of the disease process's molecular mechanisms will facilitate development of highly effective diagnostic and therapeutic treatments. Such insights can also inform preventive strategies to minimize the disease's future development.
Collapse
Affiliation(s)
- Samar A. Zailaie
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Basmah B. Khoja
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Jumana J. Siddiqui
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Mohammad H. Mawardi
- Medicine Department, Gastroenterology Section, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia;
| | - Emily Heaphy
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Amjad Aljagthmi
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Consolato M. Sergi
- Children’s Hospital of Eastern Ontario (CHEO), University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
47
|
Lozano-Velasco E, Inácio JM, Sousa I, Guimarães AR, Franco D, Moura G, Belo JA. miRNAs in Heart Development and Disease. Int J Mol Sci 2024; 25:1673. [PMID: 38338950 PMCID: PMC10855082 DOI: 10.3390/ijms25031673] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/12/2024] Open
Abstract
Cardiovascular diseases (CVD) are a group of disorders that affect the heart and blood vessels. They include conditions such as myocardial infarction, coronary artery disease, heart failure, arrhythmia, and congenital heart defects. CVDs are the leading cause of death worldwide. Therefore, new medical interventions that aim to prevent, treat, or manage CVDs are of prime importance. MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression at the posttranscriptional level and play important roles in various biological processes, including cardiac development, function, and disease. Moreover, miRNAs can also act as biomarkers and therapeutic targets. In order to identify and characterize miRNAs and their target genes, scientists take advantage of computational tools such as bioinformatic algorithms, which can also assist in analyzing miRNA expression profiles, functions, and interactions in different cardiac conditions. Indeed, the combination of miRNA research and bioinformatic algorithms has opened new avenues for understanding and treating CVDs. In this review, we summarize the current knowledge on the roles of miRNAs in cardiac development and CVDs, discuss the challenges and opportunities, and provide some examples of recent bioinformatics for miRNA research in cardiovascular biology and medicine.
Collapse
Affiliation(s)
- Estefania Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (D.F.)
| | - José Manuel Inácio
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal;
| | - Inês Sousa
- Genome Medicine Lab, Department of Medical Sciences, Institute for Biomedicine–iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal; (I.S.); (A.R.G.); (G.M.)
| | - Ana Rita Guimarães
- Genome Medicine Lab, Department of Medical Sciences, Institute for Biomedicine–iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal; (I.S.); (A.R.G.); (G.M.)
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (D.F.)
| | - Gabriela Moura
- Genome Medicine Lab, Department of Medical Sciences, Institute for Biomedicine–iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal; (I.S.); (A.R.G.); (G.M.)
| | - José António Belo
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal;
| |
Collapse
|
48
|
Ha Thi HT, Than VT. Recent applications of RNA therapeutic in clinics. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 203:115-150. [PMID: 38359994 DOI: 10.1016/bs.pmbts.2023.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Ribonucleic acid (RNA) therapy has been extensively researched for several decades and has garnered significant attention in recent years owing to its potential in treating a broad spectrum of diseases. It falls under the domain of gene therapy, leveraging RNA molecules as a therapeutic approach in medicine. RNA can be targeted using small-molecule drugs, or RNA molecules themselves can serve as drugs by interacting with proteins or other RNA molecules. While several RNA drugs have been granted clinical approval, numerous RNA-based therapeutics are presently undergoing clinical investigation or testing for various conditions, including genetic disorders, viral infections, and diverse forms of cancer. These therapies offer several advantages, such as high specificity, enabling precise targeting of disease-related genes or proteins, cost-effectiveness, and a relatively straightforward manufacturing process. Nevertheless, successful translation of RNA therapies into widespread clinical use necessitates addressing challenges related to delivery, stability, and potential off-target effects. This chapter provides a comprehensive overview of the general concepts of various classes of RNA-based therapeutics, the mechanistic basis of their function, as well as recent applications of RNA therapeutic in clinics.
Collapse
Affiliation(s)
- Huyen Trang Ha Thi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea.
| | - Van Thai Than
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam; Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
| |
Collapse
|
49
|
Shrivastav D, Singh DD. Emerging roles of microRNAs as diagnostics and potential therapeutic interest in type 2 diabetes mellitus. World J Clin Cases 2024; 12:525-537. [PMID: 38322458 PMCID: PMC10841963 DOI: 10.12998/wjcc.v12.i3.525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/18/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a metabolic disease of impaired glucose utilization. Uncontrolled high sugar levels lead to advanced glycation end products (AGEs), which affects several metabolic pathways by its receptor of advanced glycation end products (RAGE) and causes diabetic complication. MiRNAs are small RNA molecules which regulate genes linked to diabetes and affect AGEs pathogenesis, and target tissues, influencing health and disease processes. AIM To explore miRNA roles in T2DM's metabolic pathways for potential therapeutic and diagnostic advancements in diabetes complications. METHODS We systematically searched the electronic database PubMed using keywords. We included free, full-length research articles that evaluate the role of miRNAs in T2DM and its complications, focusing on genetic and molecular disease mechanisms. After assessing the full-length papers of the shortlisted articles, we included 12 research articles. RESULTS Several types of miRNAs are linked in metabolic pathways which are affected by AGE/RAGE axis in T2DM and its complications. miR-96-5p, miR-7-5p, miR-132, has_circ_0071106, miR-143, miR-21, miR-145-5p, and more are associated with various aspects of T2DM, including disease risk, diagnostic markers, complications, and gene regulation. CONCLUSION Targeting the AGE/RAGE axis, with a focus on miRNA regulation, holds promise for managing T2DM and its complications. MiRNAs have therapeutic potential as they can influence the metabolic pathways affected by AGEs and RAGE, potentially reducing inflammation, oxidative stress, and vascular complications. Additionally, miRNAs may serve as early diagnostic biomarkers for T2DM. Further research in this area may lead to innovative therapeutic strategies for diabetes and its associated complications.
Collapse
Affiliation(s)
| | - Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur 303002, India
| |
Collapse
|
50
|
Le Huy B, Bui Thi Phuong H, Luong Xuan H. Advantages and disadvantages of RNA therapeutics. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 203:151-164. [PMID: 38359996 DOI: 10.1016/bs.pmbts.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
RNA therapeutics is an innovative and rapidly evolving field at the forefront of medical research and biotechnology. Recently, many studies have shown that diverse RNA types play important roles in cells. Besides the protein translation coding, they also express and regulate a variety of cellular pathways. Indeed, along with the research and studies, many drugs and vaccines were developed from RNAs, including both coding and non-coding RNA. Some cases were approved to be medicines or under clinical trial. After years of use and application, they have shown a bright opportunity to prevent and treat many fatal and rare diseases with many strong points, such as fast production and long-term effects. Besides, they still have some drawbacks that need to be overcome, like stability or delivery to become the new generation of medicine. Therefore, this chapter focuses on providing an overview of the advantages and disadvantages of RNA therapeutics as well as some crucial points for future development.
Collapse
Affiliation(s)
- Binh Le Huy
- Center for High Technology Development, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam; School of Chemical Engineering-Hanọi University of Science and Technology, Hanoi, Vietnam
| | | | - Huy Luong Xuan
- Faculty of Pharmacy, PHENIKAA University, Hanoi, Vietnam; PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi, Vietnam.
| |
Collapse
|