1
|
Qin J, Tan Y, Han Y, Yu L, Liu S, Zhao S, Wan H, Qu S. Interplay Between TGF-β Signaling and MicroRNA in Diabetic Cardiomyopathy. Cardiovasc Drugs Ther 2025; 39:633-641. [PMID: 38117422 DOI: 10.1007/s10557-023-07532-2] [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] [Accepted: 11/20/2023] [Indexed: 12/21/2023]
Abstract
In diabetic patients, concomitant cardiovascular disease is the main factor contributing to their morbidity and mortality. Diabetic cardiomyopathy (DCM) is a form of cardiovascular disease associated with diabetes that can result in heart failure. Transforming growth factor-β (TGF-β) isoforms play a crucial role in heart remodeling and repair and are elevated and activated in myocardial disorders. Alterations in certain microRNAs (miRNA) are closely related to diabetic cardiomyopathy. One or more miRNA molecules target the majority of TGF-β pathway components, and TGF-β directly or via SMADs controls miRNA synthesis. Based on these interactions, this review discusses potential cross-talk between TGF-β signaling and miRNA in DCM in order to investigate the creation of potential therapeutic targets.
Collapse
Affiliation(s)
- Jianning Qin
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hongxiang Street, Hengyang, 421001, Hunan, China
| | - Yao Tan
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hongxiang Street, Hengyang, 421001, Hunan, China
| | - Yang Han
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hongxiang Street, Hengyang, 421001, Hunan, China
| | - Letian Yu
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hongxiang Street, Hengyang, 421001, Hunan, China
| | - Shali Liu
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hongxiang Street, Hengyang, 421001, Hunan, China
| | - Simin Zhao
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hongxiang Street, Hengyang, 421001, Hunan, China
| | - Hengquan Wan
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hongxiang Street, Hengyang, 421001, Hunan, China
| | - Shunlin Qu
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hongxiang Street, Hengyang, 421001, Hunan, China.
| |
Collapse
|
2
|
Chen J, Luo M, Xing Z, Chen Y, Peng C, Li D. Start small, think big: MicroRNAs in diabetes mellitus and relevant cardiorenal-liver metabolic health spectrum. Metabolism 2025; 165:156153. [PMID: 39914482 DOI: 10.1016/j.metabol.2025.156153] [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: 08/29/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Diabetes mellitus (DM), co-existing with metabolic disorder of cardio-renal-liver, is one of the most difficult problems in medicine that attracts global concern with high mortality. MicroRNAs (miRNAs) are a class of small non-coding RNA molecules that negatively regulates gene expression and exerts active against a large proportion of the transcriptome, due to their high evolutionary conservation. Emerging evidence prove that miRNAs are involved in the pathogenesis of DM and associated metabolic disorders, manifested by their variable alteration in the blood, urine, tissues, or organs, principally contributing to modulate the interconnections between DM and cardio-renal-liver metabolism. Mechanistically, miRNAs regulate various biological processes, such as metabolism of insulin, lipid, glucose, inflammatory response, fibrosis, oxidative stress, apoptosis, and angiogenesis, etc. This review emphasizes the function of miRNAs and highlights the physiopathological regulation of miRNA in DM and related complications, especially the dysfunction of cardiovascular system, kidneys, and liver, with the aim of providing promising biomarkers for assisting early diagnosis of DM with cardio-renal-liver- specific metabolic disorders, as well as for the development of miRNA-targeting agents.
Collapse
Affiliation(s)
- Junren Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maozhu Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziwei Xing
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
3
|
Kura B, Kindernay L, Singla D, Dulova U, Bartekova M. Mechanistic insight into the role of cardiac-enriched microRNAs in diabetic heart injury. Am J Physiol Heart Circ Physiol 2025; 328:H865-H884. [PMID: 40033927 PMCID: PMC12069993 DOI: 10.1152/ajpheart.00736.2024] [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: 10/24/2024] [Revised: 11/12/2024] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Cardiovascular complications, particularly diabetic cardiomyopathy (DCM), are the primary causes of morbidity and mortality among individuals with diabetes. Hyperglycemia associated with diabetes leads to cardiomyocyte hypertrophy, apoptosis, and myocardial fibrosis, culminating in heart failure (HF). Patients with diabetes face a 2-4 times greater risk of developing HF compared with those without diabetes. Consequently, there is a growing interest in exploring the molecular mechanisms that contribute to the development of DCM. MicroRNAs (miRNAs) are short, single-stranded, noncoding RNA molecules that participate in the maintenance of physiological homeostasis through the regulation of essential processes such as metabolism, cell proliferation, and apoptosis. At the posttranscriptional level, miRNAs modulate gene expression by binding directly to genes' mRNAs. Multiple cardiac-enriched miRNAs were reported to be dysregulated under diabetic conditions. Different studies revealed the role of specific miRNAs in the pathogenesis of diabetes and related cardiovascular complications, including cardiomyocyte hypertrophy and fibrosis, mitochondrial dysfunction, metabolic impairment, inflammatory response, or cardiomyocyte death. Circulating miRNAs have been shown to represent the potential biomarkers for early detection of diabetic heart injury. A deeper understanding of miRNAs and their role in diabetes-related pathophysiological processes could lead to new therapeutic strategies for addressing cardiac complications associated with diabetes.
Collapse
Affiliation(s)
- Branislav Kura
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lucia Kindernay
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Dinender Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, USA
| | - Ulrika Dulova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
4
|
Nueraihemaiti N, Dilimulati D, Baishan A, Hailati S, Maihemuti N, Aikebaier A, Paerhati Y, Zhou W. Shared Genomic Features Between Lung Adenocarcinoma and Type 2 Diabetes: A Bioinformatics Study. BIOLOGY 2025; 14:331. [PMID: 40282196 PMCID: PMC12025072 DOI: 10.3390/biology14040331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a common histopathological variant of non-small cell lung cancer. Individuals with type 2 diabetes (T2DM) face an elevated risk of developing LUAD. We examined the common genomic characteristics between LUAD and T2DM through bioinformatics analysis. METHODS We acquired the GSE40791, GSE25724, GSE10072, and GSE71416 datasets. Differentially expressed genes (DEGs) were identified through R software, particularly its version 4.1.3 and analyzed via gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Subsequently, we analyzed the relationship between immune cell infiltration and DEGs. we constructed a protein-protein interaction network using STRING and visualized it with Cytoscape. Moreover, gene modules were identified utilizing the MCODE plugin, and hub genes were selected through the CytoHubba plugin. Additionally, we evaluated the predictive significance of hub genes using receiver operating characteristic curves and identified the final central hub genes. Finally, we forecasted the regulatory networks of miRNA and transcription factors for the central hub genes. RESULTS A total of 748 DEGs were identified. Analysis of immune infiltration showed a notable accumulation of effector-memory CD8 T cells, T follicular helper cells, type 1 T helper cells, activated B cells, natural killer cells, macrophages, and neutrophils in both LUAD and T2DM. Moreover, these DEGs were predominantly enriched in immune-related pathways, including the positive regulation of I-κB kinase/NF-κB signaling, positive regulation of immunoglobulin production, cellular response to interleukin-7, and cellular response to interleukin-4. The TGF-β signaling pathway was significantly important among them. Additionally, seven hub genes were identified, including ATR, RFC4, MCM2, NUP155, NUP107, NUP85, and NUP37. Among them, ATR, RFC4, and MCM2 were identified as pivotal hub genes. Additionally, hsa-mir147a, hsa-mir16-5p, and hsa-mir-1-3p were associated with LUAD and T2DM. SP1 (specific protein 1) and KDM5A (lysine-specific demethylase 5A) regulated MCM2, ATR, and RFC4. CONCLUSIONS Our study elucidates the common mechanisms of immune response, TGF-β signaling pathway, and natural killer cells in LUAD and T2DM, and identifies ATR, RFC4, and MCM2 as key potential biomarkers and therapeutic targets for the comorbidity of these two conditions.
Collapse
Affiliation(s)
- Nuerbiye Nueraihemaiti
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Dilihuma Dilimulati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Alhar Baishan
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Sendaer Hailati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Nulibiya Maihemuti
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Alifeiye Aikebaier
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Yipaerguli Paerhati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Wenting Zhou
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| |
Collapse
|
5
|
Balan AI, Scridon A. MicroRNAs in atrial fibrillation - have we discovered the Holy Grail or opened a Pandora's box? Front Pharmacol 2025; 16:1535621. [PMID: 40012622 PMCID: PMC11861496 DOI: 10.3389/fphar.2025.1535621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025] Open
Abstract
Atrial fibrillation (AF) causes a heavy socio-economic burden on healthcare systems around the globe. Identification of new preventive, diagnostic, and treatment methods is imperative. In recent years, special attention has been paid to microRNAs (miRNAs) as potential regulators of AF pathogenesis. Through post-transcriptional regulation of genes, miRNAs have been shown to play crucial roles in AF-related structural and electrical atrial remodeling. Altered expression of different miRNAs has been related to proarrhythmic changes in the duration of action potentials and atrial fibrosis. In clinical studies, miRNA changes have been associated with AF, whereas in experimental studies miRNA manipulation has emerged as a potential therapeutic approach. It would appear that, with the advent of miRNAs, we may have found the Holy Grail, and that efficient and personalized AF therapy may be one step away. Yet, the clinical relevance of miRNA evaluation and manipulation remains questionable. Studies have identified numerous miRNAs associated with AF, but none of them have shown sufficient specificity for AF. MicroRNAs are not gene-specific but regulate the expression of a myriad of genes. Cardiac and non-cardiac off-target effects may thus occur following miRNA manipulation. A Pandora's box might thus have opened with the advent of these sophisticated molecules. In this paper, we provide a critical analysis of the clinical and experimental, epidemiological and mechanistic data linking miRNAs to AF, we discuss the most promising miRNA therapeutic approaches, we emphasize a number of questions that remain to be answered, and we identify hotspots for future research.
Collapse
Affiliation(s)
| | - Alina Scridon
- Physiology Department and Center for Advanced Medical and Pharmaceutical Research, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, University of Medicine, Târgu Mures, Romania
| |
Collapse
|
6
|
Khedr NF, Zahran ES, Ebeid AM, Melek ST, Werida RH. Effect of green coffee on miR-133a, miR-155 and inflammatory biomarkers in obese individuals. Diabetol Metab Syndr 2024; 16:256. [PMID: 39468643 PMCID: PMC11520395 DOI: 10.1186/s13098-024-01478-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/23/2024] [Indexed: 10/30/2024] Open
Abstract
OBJECTIVES Metabolic syndrome is a cluster of conditions that increases the risk of atherosclerotic cardiovascular diseases. The current study was a randomized, double blind, placebo-controlled study that aimed to determine the impact of green coffee (GC) in obese patients with metabolic syndrome through analysis of miRNA-155, miRNA-133a and the inflammatory biomarkers such as resistin, TNF-α, total sialic acid, homocysteine, high sensitivity C-reactive protein (hs-CRP), and the anti-inflammatory cytokine, adiponectin. METHODS One hundred-sixty obese patients were randomly supplemented either with GC capsules (800 mg) or placebo daily for six months. Both groups were advised to take a balanced diet. Blood samples were collected at baseline and after six months of supplementation. RESULTS GC supplementation for 6 months reduced BMI (p = 0.002), waist circumference (p = 0.038), blood glucose (p = 0.002), HbA1c% (p = 0.000), Insulin (p = 0.000), systolic blood pressure (p = 0.005), diastolic BP (p = 0.001) compared with placebo. GC significantly decreased total cholesterol (TC, p = 0.000), LDL-C (p = 0.001), triglycerides (TG, p = 0.002) and increased HDL-C (p = 0.008) compared with placebo group. In addition, GC significantly (p ≤ 0.005) reduced total sialic acid, homocysteine, resistin, TNF-α, hs-CRP and the oxidative stress marker malondialdehyde (MDA), but increased serum adiponectin (p = 0.000) compared to placebo group. There was a significant reduction in the gene expression of miR-133a (p = 0.000) in GC group as compared with baseline levels and with the control placebo group (p = 0.001) after 6 months. CONCLUSION GC administration modulated metabolic syndrome by decreasing BMI, high BP, blood glucose, dyslipidemia, miRNA-133a and inflammatory biomarkers that constitute risk factors for cardiovascular diseases. CLINICALTRIALS gov registration No. is NCT05688917.
Collapse
Affiliation(s)
- Naglaa F Khedr
- Biochemistry Department, Faculty of Pharmacy, Medical Complex, Tanta University, Al-Baher St, Tanta, 31527, Egypt.
| | - Enas S Zahran
- Internal Medicine Department, Faculty of Medicine, Menoufia University, Shebeen Elkom, Egypt
| | - Abla M Ebeid
- Clinical Pharmacy Department, Faculty of Pharmacy, AL-Delta University, Gamasa, Egypt
| | - Samuel T Melek
- Department of Parasitology and Blood Research at National Organization for Drug Control and Research (NODCAR), 12654, Cairo, Egypt
| | - Rehab H Werida
- Clinical Pharmacy Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt.
| |
Collapse
|
7
|
Moghadasi M, Taherimoghaddam M, Babaeenezhad E, Birjandi M, Kaviani M, Moradi Sarabi M. MicroRNA-34a and promoter methylation contribute to peroxisome proliferator-activated receptor gamma gene expression in patients with type 2 diabetes. Diabetes Metab Syndr 2024; 18:103156. [PMID: 39522431 DOI: 10.1016/j.dsx.2024.103156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
AIMS This study aimed to investigate the roles of DNA methylation and miR-34a in the regulation of peroxisome proliferator-activated receptor gamma (PPARγ) in patients with type 2 diabetes (T2D). METHODS We investigated the methylation status of four regions of the PPARγ promoter and PPARγ expression in a panel of 84 T2D patients using methylation-specific PCR (MSP) and RT-qPCR, respectively. Moreover, we quantified DNA methyltransferases (DNMTs) expression and global DNA methylation levels by RT-qPCR and ELISA, respectively. We measured the expression levels of miR-34a and protein expression of PPARγ by stem-loop RT-qPCR and ELISA, respectively. RESULTS We found significant DNA hypermethylation in the R2 and R3 regions of the PPARγ promoter in people with diabetes. Functionally, this was associated with a significant reduction in PPARγ expression. In addition, we observed a significant increase in 5-methylcytosine levels in people with diabetes. A marked increase in circulating miR-34a in the early stages of T2D (up to 10 years) and a significant decrease in circulating miR-34a with increasing diabetes duration from 10 years after the onset of diabetes. Interestingly, upregulation of DNA methyltransferases 1 (DNMT1), DNMT3A, and DNMT3B was observed in people with diabetes, and the average expression of DNMTs was negatively correlated with circulating miR-34a levels. In contrast, the serum protein level of PPARγ, a direct target of miR-34a, increased considerably with diabetes duration and showed a negative correlation with circulating miR-34a, cholesterol, triglyceride, and low-density lipoprotein. CONCLUSION PPARγ promoter hypermethylation and miR-34a upregulation are associated with T2D pathogenesis through PPARγ dysregulation.
Collapse
Affiliation(s)
- Mona Moghadasi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Department Clinical Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mozhgan Taherimoghaddam
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Department Clinical Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Esmaeel Babaeenezhad
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Department Clinical Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mehdi Birjandi
- Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Department of Biostatistics and Epidemiology, School of Health and Nutrition, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mozhgan Kaviani
- Department of Internal Medicine, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mostafa Moradi Sarabi
- Department Clinical Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran; Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
8
|
Elsayed AK, Aldous N, Alajez NM, Abdelalim EM. Identifying miRNA Signatures Associated with Pancreatic Islet Dysfunction in a FOXA2-Deficient iPSC Model. Stem Cell Rev Rep 2024; 20:1915-1931. [PMID: 38916841 PMCID: PMC11445299 DOI: 10.1007/s12015-024-10752-0] [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] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
The pathogenesis of diabetes involves complex changes in the expression profiles of mRNA and non-coding RNAs within pancreatic islet cells. Recent progress in induced pluripotent stem cell (iPSC) technology have allowed the modeling of diabetes-associated genes. Our recent study using FOXA2-deficient human iPSC models has highlighted an essential role for FOXA2 in the development of human pancreas. Here, we aimed to provide further insights on the role of microRNAs (miRNAs) by studying the miRNA-mRNA regulatory networks in iPSC-derived islets lacking the FOXA2 gene. Consistent with our previous findings, the absence of FOXA2 significantly downregulated the expression of islet hormones, INS, and GCG, alongside other key developmental genes in pancreatic islets. Concordantly, RNA-Seq analysis showed significant downregulation of genes related to pancreatic development and upregulation of genes associated with nervous system development and lipid metabolic pathways. Furthermore, the absence of FOXA2 in iPSC-derived pancreatic islets resulted in significant alterations in miRNA expression, with 61 miRNAs upregulated and 99 downregulated. The upregulated miRNAs targeted crucial genes involved in diabetes and pancreatic islet cell development. In contrary, the absence of FOXA2 in islets showed a network of downregulated miRNAs targeting genes related to nervous system development and lipid metabolism. These findings highlight the impact of FOXA2 absence on pancreatic islet development and suggesting intricate miRNA-mRNA regulatory networks affecting pancreatic islet cell development.
Collapse
Affiliation(s)
- Ahmed K Elsayed
- Laboratory of Pluripotent Stem Cell Disease Modeling, Translational Medicine Department, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar
- Stem Cell Core, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Noura Aldous
- Laboratory of Pluripotent Stem Cell Disease Modeling, Translational Medicine Department, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Nehad M Alajez
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Essam M Abdelalim
- Laboratory of Pluripotent Stem Cell Disease Modeling, Translational Medicine Department, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar.
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar.
| |
Collapse
|
9
|
Letonja J, Petrovič D. A Review of MicroRNAs and lncRNAs in Atherosclerosis as Well as Some Major Inflammatory Conditions Affecting Atherosclerosis. Biomedicines 2024; 12:1322. [PMID: 38927529 PMCID: PMC11201627 DOI: 10.3390/biomedicines12061322] [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: 04/25/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
It is generally accepted that atherosclerosis is a chronic inflammatory disease. The link between atherosclerosis and other inflammatory diseases such as psoriasis, type 2 diabetes mellitus (T2DM), and rheumatoid arthritis (RA) via metabolic, inflammatory, and immunoregulatory pathways is well established. The aim of our review was to summarize the associations between selected microRNAs (miRs) and long non-coding RNAs (lncRNAs) and atherosclerosis, psoriasis, T2DM, and RA. We reviewed the role of miR-146a, miR-210, miR-143, miR-223, miR-126, miR-21, miR-155, miR-145, miR-200, miR-133, miR-135, miR-221, miR-424, let-7, lncRNA-H19, lncRNA-MEG3, lncRNA-UCA1, and lncRNA-XIST in atherosclerosis and psoriasis, T2DM, and RA. Extracellular vesicles (EVs) are a method of intracellular signal transduction. Their function depends on surface expression, cargo, and the cell from which they originate. The majority of the studies that investigated lncRNAs and some miRs had relatively small sample sizes, which limits the generalizability of their findings and indicates the need for more research. Based on the studies reviewed, miR-146a, miR-155, miR-145, miR-200, miR-133, and lncRNA-H19 are the most promising potential biomarkers and, possibly, therapeutic targets for atherosclerosis as well as T2DM, RA, and psoriasis.
Collapse
Affiliation(s)
- Jernej Letonja
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia;
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| | - Danijel Petrovič
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia;
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| |
Collapse
|
10
|
He R, Chen Y. The Role of Adipose Tissue-derived Exosomes in Chronic Metabolic Disorders. Curr Med Sci 2024; 44:463-474. [PMID: 38900388 DOI: 10.1007/s11596-024-2902-2] [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: 11/08/2023] [Accepted: 05/28/2024] [Indexed: 06/21/2024]
Abstract
Excessive fat deposition in obese subjects promotes the occurrence of metabolic diseases, such as type 2 diabetes mellitus (T2DM), cardiovascular diseases, and non-alcoholic fatty liver disease (NAFLD). Adipose tissue is not only the main form of energy storage but also an endocrine organ that not only secretes adipocytokines but also releases many extracellular vesicles (EVs) that play a role in the regulation of whole-body metabolism. Exosomes are a subtype of EVs, and accumulating evidence indicates that adipose tissue exosomes (AT Exos) mediate crosstalk between adipose tissue and multiple organs by being transferred to targeted cells or tissues through paracrine or endocrine mechanisms. However, the roles of AT Exos in crosstalk with metabolic organs remain to be fully elucidated. In this review, we summarize the latest research progress on the role of AT Exos in the regulation of metabolic disorders. Moreover, we discuss the potential role of AT Exos as biomarkers in metabolic diseases and their clinical application.
Collapse
Affiliation(s)
- Rui He
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Laboratory of Endocrinology & Metabolism, Key Laboratory of Vascular Aging of the Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yong Chen
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Laboratory of Endocrinology & Metabolism, Key Laboratory of Vascular Aging of the Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, 430030, China.
| |
Collapse
|
11
|
Malin SK, Erdbrügger U. Extracellular Vesicles in Metabolic and Vascular Insulin Resistance. J Vasc Res 2024; 61:129-141. [PMID: 38615667 PMCID: PMC11149383 DOI: 10.1159/000538197] [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: 12/20/2023] [Accepted: 03/01/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Insulin resistance is a major etiological factor in obesity, type 2 diabetes, and cardiovascular disease (CVD). Endothelial dysfunction may precede impairments in insulin-stimulated glucose uptake, thereby making it a key feature in development of CVD. However, the mechanism by which vascular tissue becomes dysfunctional is not clear. SUMMARY Extracellular vesicles (EVs) have emerged as potential mediators of insulin resistance and vascular dysfunction. EVs are membrane-bound particles released by tissues following cellular stress or activation. They carry "cargo" (e.g., insulin signaling proteins, eNOS-nitric oxide, and miRNA) that are believed to promote inter-cellular and interorgan communications. Herein, we review the underlying physiology of EVs in relation to type 2 diabetes and CVD risk. Specifically, we discuss how EVs may modulate metabolic (e.g., skeletal muscle, liver, and adipose) insulin sensitivity, and propose that EVs may modulate vascular insulin action to influence both endothelial function and arterial stiffness. We lastly identify how EVs may play a unique role following exercise to promote metabolic and vascular insulin sensitivity changes. KEY MESSAGE Gaining insight toward insulin-mediated EV mechanism has potential to identify novel pathways regulating cardiometabolic health and provide foundation for examining EVs as unique biomarkers and targets to prevent and/or treat chronic diseases.
Collapse
Affiliation(s)
- Steven K. Malin
- Department of Kinesiology & Health, Rutgers University, New Brunswick, NJ
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, New Brunswick, NJ
- The New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ
- Institute of Translational Medicine & Science, Rutgers University, New Brunswick, NJ
| | - Uta Erdbrügger
- Division of Nephrology, Department of Medicine, University of Virginia Health System, VA
| |
Collapse
|
12
|
Abu-Toamih-Atamni HJ, Lone IM, Binenbaum I, Mott R, Pilalis E, Chatziioannou A, Iraqi FA. Mapping novel QTL and fine mapping of previously identified QTL associated with glucose tolerance using the collaborative cross mice. Mamm Genome 2024; 35:31-55. [PMID: 37978084 DOI: 10.1007/s00335-023-10025-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/08/2023] [Indexed: 11/19/2023]
Abstract
A chronic metabolic illness, type 2 diabetes (T2D) is a polygenic and multifactorial complicated disease. With an estimated 463 million persons aged 20 to 79 having diabetes, the number is expected to rise to 700 million by 2045, creating a significant worldwide health burden. Polygenic variants of diabetes are influenced by environmental variables. T2D is regarded as a silent illness that can advance for years before being diagnosed. Finding genetic markers for T2D and metabolic syndrome in groups with similar environmental exposure is therefore essential to understanding the mechanism of such complex characteristic illnesses. So herein, we demonstrated the exclusive use of the collaborative cross (CC) mouse reference population to identify novel quantitative trait loci (QTL) and, subsequently, suggested genes associated with host glucose tolerance in response to a high-fat diet. In this study, we used 539 mice from 60 different CC lines. The diabetogenic effect in response to high-fat dietary challenge was measured by the three-hour intraperitoneal glucose tolerance test (IPGTT) test after 12 weeks of dietary challenge. Data analysis was performed using a statistical software package IBM SPSS Statistic 23. Afterward, blood glucose concentration at the specific and between different time points during the IPGTT assay and the total area under the curve (AUC0-180) of the glucose clearance was computed and utilized as a marker for the presence and severity of diabetes. The observed AUC0-180 averages for males and females were 51,267.5 and 36,537.5 mg/dL, respectively, representing a 1.4-fold difference in favor of females with lower AUC0-180 indicating adequate glucose clearance. The AUC0-180 mean differences between the sexes within each specific CC line varied widely within the CC population. A total of 46 QTL associated with the different studied phenotypes, designated as T2DSL and its number, for Type 2 Diabetes Specific Locus and its number, were identified during our study, among which 19 QTL were not previously mapped. The genomic interval of the remaining 27 QTL previously reported, were fine mapped in our study. The genomic positions of 40 of the mapped QTL overlapped (clustered) on 11 different peaks or close genomic positions, while the remaining 6 QTL were unique. Further, our study showed a complex pattern of haplotype effects of the founders, with the wild-derived strains (mainly PWK) playing a significant role in the increase of AUC values.
Collapse
Affiliation(s)
- Hanifa J Abu-Toamih-Atamni
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, 69978, Tel-Aviv, Israel
| | - Iqbal M Lone
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, 69978, Tel-Aviv, Israel
| | - Ilona Binenbaum
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Soranou Ephessiou Str, 11527, Athens, Greece
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Richard Mott
- Department of Genetics, University College of London, London, UK
| | | | - Aristotelis Chatziioannou
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Soranou Ephessiou Str, 11527, Athens, Greece
- e-NIOS Applications PC, 196 Syggrou Ave., 17671, Kallithea, Greece
| | - Fuad A Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, 69978, Tel-Aviv, Israel.
| |
Collapse
|
13
|
Caño-Carrillo S, Castillo-Casas JM, Franco D, Lozano-Velasco E. Unraveling the Signaling Dynamics of Small Extracellular Vesicles in Cardiac Diseases. Cells 2024; 13:265. [PMID: 38334657 PMCID: PMC10854837 DOI: 10.3390/cells13030265] [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: 12/29/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024] Open
Abstract
Effective intercellular communication is essential for cellular and tissue balance maintenance and response to challenges. Cellular communication methods involve direct cell contact or the release of biological molecules to cover short and long distances. However, a recent discovery in this communication network is the involvement of extracellular vesicles that host biological contents such as proteins, nucleic acids, and lipids, influencing neighboring cells. These extracellular vesicles are found in body fluids; thus, they are considered as potential disease biomarkers. Cardiovascular diseases are significant contributors to global morbidity and mortality, encompassing conditions such as ischemic heart disease, cardiomyopathies, electrical heart diseases, and heart failure. Recent studies reveal the release of extracellular vesicles by cardiovascular cells, influencing normal cardiac function and structure. However, under pathological conditions, extracellular vesicles composition changes, contributing to the development of cardiovascular diseases. Investigating the loading of molecular cargo in these extracellular vesicles is essential for understanding their role in disease development. This review consolidates the latest insights into the role of extracellular vesicles in diagnosis and prognosis of cardiovascular diseases, exploring the potential applications of extracellular vesicles in personalized therapies, shedding light on the evolving landscape of cardiovascular medicine.
Collapse
Affiliation(s)
| | | | | | - Estefanía Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (S.C.-C.); (J.M.C.-C.); (D.F.)
| |
Collapse
|
14
|
Gao S, Dong Y, Yan C, Yu T, Cao H. The role of exosomes and exosomal microRNA in diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2024; 14:1327495. [PMID: 38283742 PMCID: PMC10811149 DOI: 10.3389/fendo.2023.1327495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/27/2023] [Indexed: 01/30/2024] Open
Abstract
Diabetic cardiomyopathy, a formidable cardiovascular complication linked to diabetes, is witnessing a relentless surge in its incidence. Despite extensive research efforts, the primary pathogenic mechanisms underlying this condition remain elusive. Consequently, a critical research imperative lies in identifying a sensitive and dependable marker for early diagnosis and treatment, thereby mitigating the onset and progression of diabetic cardiomyopathy (DCM). Exosomes (EXOs), minute vesicles enclosed within bilayer lipid membranes, have emerged as a fascinating frontier in this quest, capable of transporting a diverse cargo that mirrors the physiological and pathological states of their parent cells. These exosomes play an active role in the intercellular communication network of the cardiovascular system. Within the realm of exosomes, MicroRNA (miRNA) stands as a pivotal molecular player, revealing its profound influence on the progression of DCM. This comprehensive review aims to offer an introductory exploration of exosome structure and function, followed by a detailed examination of the intricate role played by exosome-associated miRNA in diabetic cardiomyopathy. Our ultimate objective is to bolster our comprehension of DCM diagnosis and treatment strategies, thereby facilitating timely intervention and improved outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Hongbo Cao
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
15
|
Du Y, Wu L, Wang L, Reiter RJ, Lip GYH, Ren J. Extracellular vesicles in cardiovascular diseases: From pathophysiology to diagnosis and therapy. Cytokine Growth Factor Rev 2023; 74:40-55. [PMID: 37798169 DOI: 10.1016/j.cytogfr.2023.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023]
Abstract
Extracellular vesicles (EVs), encompassing exosomes, microvesicles (MVs), and apoptotic bodies (ABs), are cell-derived heterogeneous nanoparticles with a pivotal role in intercellular communication. EVs are enclosed by a lipid-bilayer membrane to escape enzymatic degradation. EVs contain various functional molecules (e.g., nucleic acids, proteins, lipids and metabolites) which can be transferred from donor cells to recipient cells. EVs provide many advantages including accessibility, modifiability and easy storage, stability, biocompatibility, heterogeneity and they readily penetrate through biological barriers, making EVs ideal and promising candidates for diagnosis/prognosis biomarkers and therapeutic tools. Recently, EVs were implicated in both physiological and pathophysiological settings of cardiovascular system through regulation of cell-cell communication. Numerous studies have reported a role for EVs in the pathophysiological progression of cardiovascular diseases (CVDs) and have evaluated the utility of EVs for the diagnosis/prognosis and therapeutics of CVDs. In this review, we summarize the biology of EVs, evaluate the perceived biological function of EVs in different CVDs along with a consideration of recent progress for the application of EVs in diagnosis/prognosis and therapies of CVDs.
Collapse
Affiliation(s)
- Yuxin Du
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Lin Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Litao Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, TX, USA
| | - Gregory Y H Lip
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Laboratory Medicine and Pathology, University of Washington, Seattle WA98195, USA.
| |
Collapse
|
16
|
Catanzaro G, Conte F, Trocchianesi S, Splendiani E, Bimonte VM, Mocini E, Filardi T, Po A, Besharat ZM, Gentile MC, Paci P, Morano S, Migliaccio S, Ferretti E. Network analysis identifies circulating miR-155 as predictive biomarker of type 2 diabetes mellitus development in obese patients: a pilot study. Sci Rep 2023; 13:19496. [PMID: 37945677 PMCID: PMC10636008 DOI: 10.1038/s41598-023-46516-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
Obesity is the main risk factor for many non-communicable diseases. In clinical practice, unspecific markers are used for the determination of metabolic alterations and inflammation, without allowing the characterization of subjects at higher risk of complications. Circulating microRNAs represent an attractive approach for early screening to identify subjects affected by obesity more at risk of developing connected pathologies. The aim of this study was the identification of circulating free and extracellular vesicles (EVs)-embedded microRNAs able to identify obese patients at higher risk of type 2 diabetes (DM2). The expression data of circulating microRNAs derived from obese patients (OB), with DM2 (OBDM) and healthy donors were combined with clinical data, through network-based methodology implemented by weighted gene co-expression network analysis. The six circulating microRNAs overexpressed in OBDM patients were evaluated in a second group of patients, confirming the overexpression of miR-155-5p in OBDM patients. Interestingly, the combination of miR-155-5p with serum levels of IL-8, Leptin and RAGE was useful to identify OB patients most at risk of developing DM2. These results suggest that miR-155-5p is a potential circulating biomarker for DM2 and that the combination of this microRNA with other inflammatory markers in OB patients can predict the risk of developing DM2.
Collapse
Affiliation(s)
- Giuseppina Catanzaro
- Department of Experimental Medicine, Sapienza University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161, Rome, Italy
| | - Federica Conte
- Institute for Systems Analysis and Computer Science "A. Ruberti" (IASI), National Research Council (CNR), 00185, Rome, Italy
| | - Sofia Trocchianesi
- Department of Experimental Medicine, Sapienza University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161, Rome, Italy
| | - Elena Splendiani
- Department of Experimental Medicine, Sapienza University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161, Rome, Italy
| | - Viviana Maria Bimonte
- Department of Movement, Human and Health Sciences, University of Foro Italico, 00135, Rome, Italy
| | - Edoardo Mocini
- Department of Experimental Medicine, Sapienza University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161, Rome, Italy
| | - Tiziana Filardi
- Department of Experimental Medicine, Sapienza University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161, Rome, Italy
| | - Agnese Po
- Department of Molecular Medicine, Sapienza University, 00161, Rome, Italy
| | - Zein Mersini Besharat
- Department of Experimental Medicine, Sapienza University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161, Rome, Italy
| | - Maria Cristina Gentile
- Department of Experimental Medicine, Sapienza University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161, Rome, Italy
| | - Paola Paci
- Department of Computer, Control and Management Engineering, Sapienza University, 00161, Rome, Italy
| | - Susanna Morano
- Department of Experimental Medicine, Sapienza University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161, Rome, Italy
| | - Silvia Migliaccio
- Department of Movement, Human and Health Sciences, University of Foro Italico, 00135, Rome, Italy.
| | - Elisabetta Ferretti
- Department of Experimental Medicine, Sapienza University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
17
|
Ma J, Bi J, Sun B, Li H, Li Y, Wang S. Zinc Improves Semen Parameters in High-Fat Diet-Induced Male Rats by Regulating the Expression of LncRNA in Testis Tissue. Biol Trace Elem Res 2023; 201:4793-4805. [PMID: 36600170 DOI: 10.1007/s12011-022-03550-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023]
Abstract
This study aimed to identify differentially expressed LncRNAs in testis tissue of male rats induced by high-fat diet and their changes after zinc supplementation, by constructing a high-fat feeding rat model, and then supplemented with zinc, and observed the expression of LncRNA in three groups of normal, high-fat fed, and zinc-intervened rats. Experimental studies show that the semen parameters of male rats with high-fat diet were decreased but recovered after zinc supplementation, and the related LncRNA also changed. Zinc may improve the high-fat diet-induced reduction of semen parameters by changing the expression of related LncRNA.
Collapse
Affiliation(s)
- Jing Ma
- Hebei Key Laboratory of Reproductive Medicine, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Street, Shijiazhuang, 050071, Xinhua District, China
| | - Jiajie Bi
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Bo Sun
- Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Huanhuan Li
- Hebei Key Laboratory of Reproductive Medicine, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Street, Shijiazhuang, 050071, Xinhua District, China
| | - Yuejia Li
- Graduate School of Hebei Medical University, Shijiazhuang, 050017, China
| | - Shusong Wang
- Hebei Key Laboratory of Reproductive Medicine, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Street, Shijiazhuang, 050071, Xinhua District, China.
- Graduate School of Chengde Medical University, Chengde, 067000, China.
- Graduate School of Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
18
|
Satyadev N, Rivera MI, Nikolov NK, Fakoya AOJ. Exosomes as biomarkers and therapy in type 2 diabetes mellitus and associated complications. Front Physiol 2023; 14:1241096. [PMID: 37745252 PMCID: PMC10515224 DOI: 10.3389/fphys.2023.1241096] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the most prevalent metabolic disorders worldwide. However, T2DM still remains underdiagnosed and undertreated resulting in poor quality of life and increased morbidity and mortality. Given this ongoing burden, researchers have attempted to locate new therapeutic targets as well as methodologies to identify the disease and its associated complications at an earlier stage. Several studies over the last few decades have identified exosomes, small extracellular vesicles that are released by cells, as pivotal contributors to the pathogenesis of T2DM and its complications. These discoveries suggest the possibility of novel detection and treatment methods. This review provides a comprehensive presentation of exosomes that hold potential as novel biomarkers and therapeutic targets. Additional focus is given to characterizing the role of exosomes in T2DM complications, including diabetic angiopathy, diabetic cardiomyopathy, diabetic nephropathy, diabetic peripheral neuropathy, diabetic retinopathy, and diabetic wound healing. This study reveals that the utilization of exosomes as diagnostic markers and therapies is a realistic possibility for both T2DM and its complications. However, the majority of the current research is limited to animal models, warranting further investigation of exosomes in clinical trials. This review represents the most extensive and up-to-date exploration of exosomes in relation to T2DM and its complications.
Collapse
Affiliation(s)
- Nihal Satyadev
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, United States
| | - Milagros I. Rivera
- University of Medicine and Health Sciences, Basseterre, St. Kitts and Nevis
| | | | | |
Collapse
|
19
|
Laura Francés J, Pagiatakis C, Di Mauro V, Climent M. Therapeutic Potential of EVs: Targeting Cardiovascular Diseases. Biomedicines 2023; 11:1907. [PMID: 37509546 PMCID: PMC10377624 DOI: 10.3390/biomedicines11071907] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/25/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Due to their different biological functions, extracellular vesicles (EVs) have great potential from a therapeutic point of view. They are released by all cell types, carrying and delivering different kinds of biologically functional cargo. Under pathological events, cells can increase their secretion of EVs and can release different amounts of cargo, thus making EVs great biomarkers as indicators of pathological progression. Moreover, EVs are also known to be able to transport and deliver cargo to different recipient cells, having an important role in cellular communication. Interestingly, EVs have recently been explored as biological alternatives for the delivery of therapeutics, being considered natural drug delivery carriers. Because cardiovascular disorders (CVDs) are the leading cause of death worldwide, in this review, we will discuss the up-to-date knowledge regarding the biophysical properties and biological components of EVs, focusing on myocardial infarction, diabetic cardiomyopathy, and sepsis-induced cardiomyopathy, three very different types of CVDs.
Collapse
Affiliation(s)
| | - Christina Pagiatakis
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Vittoria Di Mauro
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | | |
Collapse
|
20
|
Paneru BD, Hill DA. The role of extracellular vesicle-derived miRNAs in adipose tissue function and metabolic health. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00027. [PMID: 37501663 PMCID: PMC10371064 DOI: 10.1097/in9.0000000000000027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023]
Abstract
Extracellular vesicles (EVs) are nanometer size lipid particles that are released from virtually every cell type. Recent studies have shown that miRNAs carried by EVs play important roles in intercellular and interorgan communication. In the context of obesity and insulin resistance, EV-derived miRNAs functionally bridge major metabolic organs, including the adipose tissue, skeletal muscle, liver, and pancreas, to regulate insulin secretion and signaling. As a result, many of these EV-derived miRNAs have been proposed as potential disease biomarkers and/or therapeutic agents. However, the field's knowledge of EV miRNA-mediated regulation of mammalian metabolism is still in its infancy. Here, we review the evidence indicating that EV-derived miRNAs provide cell-to-cell and organ-to-organ communication to support metabolic health, highlight the potential medical relevance of these discoveries, and discuss the most important knowledge gaps and future directions for this field.
Collapse
Affiliation(s)
- Bam D. Paneru
- Division of Allergy and Immunology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - David A. Hill
- Division of Allergy and Immunology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Institute for Immunology, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
21
|
Shahabi Raberi V, Javanshir E, Abbasnezhad M, Mashayekhi S, Abbasnezhad A, Ahmadzadeh M, Shariati A. Emerging Biomarkers of Acute Myocardial Infarction, An Overview of the Newest MicroRNAs. Galen Med J 2023; 12:e2909. [PMID: 38774858 PMCID: PMC11108668 DOI: 10.31661/gmj.v12i.2909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 04/01/2025] Open
Abstract
Globally, acute myocardial infarction (AMI) is the leading cause of death. Early and precise diagnosis is essential for medical care to enhance prognoses and reduce mortality. The diagnosis of AMI relies primarily on conventional circulating biomarkers. However, these markers have many drawbacks. Non-coding RNAs (ncRNAs) form a significant fraction of the transcriptome and have been shown to be essential for many biological processes, including the pathogenesis of the disease. ncRNAs can be utilized as biomarkers due to their important role in the disease's development. The current manuscript describes recent progress on the role of ncRNAs as new AMI biomarkers.
Collapse
Affiliation(s)
- Venus Shahabi Raberi
- Seyed-Al-Shohada Cardiology Hospital, Urmia University of Medical Sciences, Urmia,
Iran
| | - Elnaz Javanshir
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Abbasnezhad
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Mashayekhi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Masumeh Ahmadzadeh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akram Shariati
- Department of Cardiology, School of Medicine, Urmia University of Medical Sciences,
Urmia, Iran
| |
Collapse
|
22
|
Shahabi Raberi V, Javanshir E, Abbasnezhad M, Mashayekhi S, Abbasnezhad A, Ahmadzadeh M, Shariati A. Emerging Biomarkers of Acute Myocardial Infarction, An Overview of the Newest MicroRNAs. Galen Med J 2023; 12:e2909. [PMID: 38774858 PMCID: PMC11108668 DOI: 10.31661/gmj.v12i0.2909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 05/24/2024] Open
Abstract
Globally, acute myocardial infarction (AMI) is the leading cause of death. Early and precise diagnosis is essential for medical care to enhance prognoses and reduce mortality. The diagnosis of AMI relies primarily on conventional circulating biomarkers. However, these markers have many drawbacks. Non-coding RNAs (ncRNAs) form a significant fraction of the transcriptome and have been shown to be essential for many biological processes, including the pathogenesis of the disease. ncRNAs can be utilized as biomarkers due to their important role in the disease's development. The current manuscript describes recent progress on the role of ncRNAs as new AMI biomarkers.
Collapse
Affiliation(s)
- Venus Shahabi Raberi
- Seyed-Al-Shohada Cardiology Hospital, Urmia University of Medical Sciences, Urmia,
Iran
| | - Elnaz Javanshir
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Abbasnezhad
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Mashayekhi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Masumeh Ahmadzadeh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akram Shariati
- Department of Cardiology, School of Medicine, Urmia University of Medical Sciences,
Urmia, Iran
| |
Collapse
|
23
|
Morales-Sánchez P, Lambert C, Ares-Blanco J, Suárez-Gutiérrez L, Villa-Fernández E, Garcia AV, García-Villarino M, Tejedor JR, Fraga MF, Torre EM, Pujante P, Delgado E. Circulating miRNA expression in long-standing type 1 diabetes mellitus. Sci Rep 2023; 13:8611. [PMID: 37244952 DOI: 10.1038/s41598-023-35836-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023] Open
Abstract
Type 1 diabetes is a chronic autoimmune disease which results in inefficient regulation of glucose homeostasis and can lead to different vascular comorbidities through life. In this study we aimed to analyse the circulating miRNA expression profile of patients with type 1 diabetes, and with no other associated pathology. For this, fasting plasma was obtained from 85 subjects. Next generation sequencing analysis was firstly performed to identify miRNAs that were differentially expressed between groups (20 patients vs. 10 controls). hsa-miR-1-3p, hsa-miR-200b-3p, hsa-miR-9-5p, and hsa-miR-1200 expression was also measured by Taqman RT-PCR to validate the observed changes (34 patients vs. 21 controls). Finally, through a bioinformatic approach, the main pathways affected by the target genes of these miRNAs were studied. Among the studied miRNAs, hsa-miR-1-3p expression was found significantly increased in patients with type 1 diabetes compared to controls, and positively correlated with glycated haemoglobin levels. Additionally, by using a bioinformatic approach, we could observe that changes in hsa-miR-1-3p directly affect genes involved in vascular development and cardiovascular pathologies. Our results suggest that, circulating hsa-miR-1-3p in plasma, together with glycaemic control, could be used as prognostic biomarkers in type 1 diabetes, helping to prevent the development of vascular complications in these patients.
Collapse
Affiliation(s)
- Paula Morales-Sánchez
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Lambert
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- University of Barcelona, Barcelona, Spain.
| | - Jessica Ares-Blanco
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
| | - Lorena Suárez-Gutiérrez
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - Elsa Villa-Fernández
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Ana Victoria Garcia
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Miguel García-Villarino
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Juan Ramón Tejedor
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Oviedo, Asturias, Spain
- Institute of Oncology of Asturias (IUOPA), Oviedo, Asturias, Spain
- Department of Organisms and Systems Biology (B.O.S), University of Oviedo, Oviedo, Asturias, Spain
| | - Mario F Fraga
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Oviedo, Asturias, Spain
- Institute of Oncology of Asturias (IUOPA), Oviedo, Asturias, Spain
- Department of Organisms and Systems Biology (B.O.S), University of Oviedo, Oviedo, Asturias, Spain
| | - Edelmiro Menéndez Torre
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
| | - Pedro Pujante
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain.
| | - Elías Delgado
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain.
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain.
| |
Collapse
|
24
|
Spinetti G, Mutoli M, Greco S, Riccio F, Ben-Aicha S, Kenneweg F, Jusic A, de Gonzalo-Calvo D, Nossent AY, Novella S, Kararigas G, Thum T, Emanueli C, Devaux Y, Martelli F. Cardiovascular complications of diabetes: role of non-coding RNAs in the crosstalk between immune and cardiovascular systems. Cardiovasc Diabetol 2023; 22:122. [PMID: 37226245 PMCID: PMC10206598 DOI: 10.1186/s12933-023-01842-3] [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: 03/31/2023] [Accepted: 04/25/2023] [Indexed: 05/26/2023] Open
Abstract
Diabetes mellitus, a group of metabolic disorders characterized by high levels of blood glucose caused by insulin defect or impairment, is a major risk factor for cardiovascular diseases and related mortality. Patients with diabetes experience a state of chronic or intermittent hyperglycemia resulting in damage to the vasculature, leading to micro- and macro-vascular diseases. These conditions are associated with low-grade chronic inflammation and accelerated atherosclerosis. Several classes of leukocytes have been implicated in diabetic cardiovascular impairment. Although the molecular pathways through which diabetes elicits an inflammatory response have attracted significant attention, how they contribute to altering cardiovascular homeostasis is still incompletely understood. In this respect, non-coding RNAs (ncRNAs) are a still largely under-investigated class of transcripts that may play a fundamental role. This review article gathers the current knowledge on the function of ncRNAs in the crosstalk between immune and cardiovascular cells in the context of diabetic complications, highlighting the influence of biological sex in such mechanisms and exploring the potential role of ncRNAs as biomarkers and targets for treatments. The discussion closes by offering an overview of the ncRNAs involved in the increased cardiovascular risk suffered by patients with diabetes facing Sars-CoV-2 infection.
Collapse
Affiliation(s)
- Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Martina Mutoli
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Federica Riccio
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| | - Soumaya Ben-Aicha
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Franziska Kenneweg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | | | - David de Gonzalo-Calvo
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Anne Yaël Nossent
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Susana Novella
- Department of Physiology, University of Valencia - INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Georgios Kararigas
- Department of Physiology, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Costanza Emanueli
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy.
| |
Collapse
|
25
|
Pérez-Cremades D, Chen J, Assa C, Feinberg MW. MicroRNA-mediated control of myocardial infarction in diabetes. Trends Cardiovasc Med 2023; 33:195-201. [PMID: 35051592 PMCID: PMC9288556 DOI: 10.1016/j.tcm.2022.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 12/16/2022]
Abstract
Diabetes mellitus is a global public health problem whose cases will continue to rise along with the progressive increase in obesity and the aging of the population. People with diabetes exhibit higher risk of cardiovascular complications, especially myocardial infarction (MI). microRNAs (miRNAs) are evolutionary conserved small non-coding RNAs involved in the regulation of biological processes by interfering in gene expression at the post-transcriptional level. Accumulating studies in the last two decades have uncovered the role of stage-specific miRNAs associated with key pathobiological events observed in the hearts of people with diabetes and MI, including cardiomyocyte death, angiogenesis, inflammatory response, myocardial remodeling, and myocardial lipotoxicity. A better understanding of the importance of these miRNAs and their targets may provide novel opportunities for RNA-based therapeutic interventions to address the increased risk of MI in diabetes.
Collapse
Affiliation(s)
- Daniel Pérez-Cremades
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA 02115; Department of Physiology, University of Valencia and INCLIVA Biomedical Research Institute, Valencia, Spain 46010
| | - Jingshu Chen
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA 02115
| | - Carmel Assa
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA 02115
| | - Mark W Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA 02115.
| |
Collapse
|
26
|
Ao X, Ding W, Li X, Xu Q, Chen X, Zhou X, Wang J, Liu Y. Non-coding RNAs regulating mitochondrial function in cardiovascular diseases. J Mol Med (Berl) 2023; 101:501-526. [PMID: 37014377 DOI: 10.1007/s00109-023-02305-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/14/2023] [Accepted: 03/13/2023] [Indexed: 04/05/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of disease-related death worldwide and a significant obstacle to improving patients' health and lives. Mitochondria are core organelles for the maintenance of myocardial tissue homeostasis, and their impairment and dysfunction are considered major contributors to the pathogenesis of various CVDs, such as hypertension, myocardial infarction, and heart failure. However, the exact roles of mitochondrial dysfunction involved in CVD pathogenesis remain not fully understood. Non-coding RNAs (ncRNAs), particularly microRNAs, long non-coding RNAs, and circular RNAs, have been shown to be crucial regulators in the initiation and development of CVDs. They can participate in CVD progression by impacting mitochondria and regulating mitochondrial function-related genes and signaling pathways. Some ncRNAs also exhibit great potential as diagnostic and/or prognostic biomarkers as well as therapeutic targets for CVD patients. In this review, we mainly focus on the underlying mechanisms of ncRNAs involved in the regulation of mitochondrial functions and their role in CVD progression. We also highlight their clinical implications as biomarkers for diagnosis and prognosis in CVD treatment. The information reviewed herein could be extremely beneficial to the development of ncRNA-based therapeutic strategies for CVD patients.
Collapse
Affiliation(s)
- Xiang Ao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, 266021, China
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Wei Ding
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Xiaoge Li
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Qingling Xu
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Xinhui Chen
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Xuehao Zhou
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Jianxun Wang
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Ying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
27
|
Sufianov A, Kostin A, Begliarzade S, Kudriashov V, Ilyasova T, Liang Y, Mukhamedzyanov A, Beylerli O. Exosomal non coding RNAs as a novel target for diabetes mellitus and its complications. Noncoding RNA Res 2023; 8:192-204. [PMID: 36818396 PMCID: PMC9929646 DOI: 10.1016/j.ncrna.2023.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Diabetes mellitus (DM) is a first-line priority among the problems facing medical science and public health in almost all countries of the world. The main problem of DM is the high incidence of damage to the cardiovascular system, which in turn leads to diseases such as myocardial infarction, stroke, gangrene of the lower extremities, blindness and chronic renal failure. As a result, the study of the molecular genetic mechanisms of the pathogenesis of DM is of critical importance for the development of new diagnostic and therapeutic strategies. Molecular genetic aspects of the etiology and pathogenesis of diabetes mellitus are intensively studied in well-known laboratories around the world. One of the strategies in this direction is to study the role of exosomes in the pathogenesis of DM. Exosomes are microscopic extracellular vesicles with a diameter of 30-100 nm, released into the intercellular space by cells of various tissues and organs. The content of exosomes depends on the cell type and includes mRNA, non-coding RNAs, DNA, and so on. Non-coding RNAs, a group of RNAs with limited transcriptional activity, have been discovered to play a significant role in regulating gene expression through epigenetic and posttranscriptional modulation, such as silencing of messenger RNA. One of the problems of usage exosomes in DM is the identification of the cellular origin of exosomes and the standardization of protocols for molecular genetic studies in clinical laboratories. In addition, the question of the target orientation of exosomes and their targeted activity requires additional study. Solving these and other problems will make it possible to use exosomes for the diagnosis and delivery of drugs directly to target cells in DM. This study presents an analysis of literature data on the role of exosomes and ncRNAs in the development and progression of DM, as well as the prospects for the use of exosomes in clinical practice in this disease.
Collapse
Affiliation(s)
- Albert Sufianov
- Educational and Scientific Institute of Neurosurgery, Рeoples’ Friendship University of Russia (RUDN University), Moscow, Russia,Department of Neurosurgery, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Andrey Kostin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia, Moscow, Russia
| | - Sema Begliarzade
- Republican Clinical Perinatal Center, Ufa, Republic of Bashkortostan, 450106, Russia
| | | | - Tatiana Ilyasova
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Yanchao Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | | | - Ozal Beylerli
- Educational and Scientific Institute of Neurosurgery, Рeoples’ Friendship University of Russia (RUDN University), Moscow, Russia,Corresponding author. Рeoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation.
| |
Collapse
|
28
|
Tarhriz V, Abkhooie L, Sarabi MM. Regulation of HIF-1 by MicroRNAs in Various Cardiovascular Diseases. Curr Cardiol Rev 2023; 19:51-56. [PMID: 37005512 PMCID: PMC10518879 DOI: 10.2174/1573403x19666230330105259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/04/2023] [Accepted: 02/07/2023] [Indexed: 04/04/2023] Open
Abstract
Today, we see an increase in death due to cardiovascular diseases all over the world, which has a lot to do with the regulation of oxygen homeostasis. Also, hypoxia-inducing factor 1 (HIF-1) is considered a vital factor in hypoxia and its physiological and pathological changes. HIF- 1 is involved in cellular activities, including proliferation, differentiation, and cell death in endothelial cells (ECs) and cardiomyocytes. Similar to HIF-1α, which acts as a protective element against various diseases in the cardiovascular system, the protective role of microRNAs (miRNAs) has also been proved using animal models. The number of miRNAs identified in the regulation of gene expression responsive to hypoxia and the importance of investigating the involvement of the non-coding genome in cardiovascular diseases is increasing, which shows the issue's importance. In this study, the molecular regulation of HIF-1 by miRNAs is considered to improve therapeutic approaches in clinical diagnoses of cardiovascular diseases.
Collapse
Affiliation(s)
- Vahideh Tarhriz
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Abkhooie
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
- Department of Medical Biotechnology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mostafa Moradi Sarabi
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
- Department of Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
29
|
Pandey A, Ajgaonkar S, Jadhav N, Saha P, Gurav P, Panda S, Mehta D, Nair S. Current Insights into miRNA and lncRNA Dysregulation in Diabetes: Signal Transduction, Clinical Trials and Biomarker Discovery. Pharmaceuticals (Basel) 2022; 15:1269. [PMID: 36297381 PMCID: PMC9610703 DOI: 10.3390/ph15101269] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/27/2022] [Accepted: 10/09/2022] [Indexed: 01/24/2023] Open
Abstract
Diabetes is one of the most frequently occurring metabolic disorders, affecting almost one tenth of the global population. Despite advances in antihyperglycemic therapeutics, the management of diabetes is limited due to its complexity and associated comorbidities, including diabetic neuropathy, diabetic nephropathy and diabetic retinopathy. Noncoding RNAs (ncRNAs), including microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are involved in the regulation of gene expression as well as various disease pathways in humans. Several ncRNAs are dysregulated in diabetes and are responsible for modulating the expression of various genes that contribute to the 'symptom complex' in diabetes. We review various miRNAs and lncRNAs implicated in diabetes and delineate ncRNA biological networks as well as key ncRNA targets in diabetes. Further, we discuss the spatial regulation of ncRNAs and their role(s) as prognostic markers in diabetes. We also shed light on the molecular mechanisms of signal transduction with diabetes-associated ncRNAs and ncRNA-mediated epigenetic events. Lastly, we summarize clinical trials on diabetes-associated ncRNAs and discuss the functional relevance of the dysregulated ncRNA interactome in diabetes. This knowledge will facilitate the identification of putative biomarkers for the therapeutic management of diabetes and its comorbidities. Taken together, the elucidation of the architecture of signature ncRNA regulatory networks in diabetes may enable the identification of novel biomarkers in the discovery pipeline for diabetes, which may lead to better management of this metabolic disorder.
Collapse
Affiliation(s)
| | | | | | - Praful Saha
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | - Pranay Gurav
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | | | - Dilip Mehta
- Synergia Life Sciences Pvt. Ltd., Mumbai 400 022, India
| | - Sujit Nair
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| |
Collapse
|
30
|
Liu H, Bahreyni A, Mohamud Y, Xue YC, Jia WW, Luo H. Enhanced genomic stability of new miRNA-regulated oncolytic coxsackievirus B3. Mol Ther Oncolytics 2022; 27:89-99. [PMID: 36321136 PMCID: PMC9593271 DOI: 10.1016/j.omto.2022.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
Abstract
Genetic modification of coxsackievirus B3 (CVB3) by inserting target sequences (TS) of tumor-suppressive and/or organ-selective microRNAs (miRs) into viral genome can efficiently eliminate viral pathogenesis without significant impacts on its oncolytic activity. Nonetheless, reversion mutants (loss of miR-TS inserts) were identified as early as day 35 post-injection in ∼40% immunodeficient mice. To improve the stability, here we re-engineered CVB3 by (1) replacing the same length of viral genome at the non-coding region with TS of cardiac-selective miR-1/miR-133 and pancreas-enriched miR-216/miR-375 or (2) inserting the above miR-TS into the coding region (i.e., P1 region) of viral genome. Serial passaging of these newly established miR-CVB3s in cultured cells for 20 rounds demonstrated significantly improved stability compared with the first-generation miR-CVB3 with 5'UTR insertion of miR-TS. The safety and stability of these new miR-CVB3s was verified in immunocompetent mice. Moreover, we showed that these new viruses retained the ability to suppress lung tumor growth in a xenograft mouse model. Finally, we observed that miR-CVB3 with insertion in P1 region was more stable than miR-CVB3 with preserved length of the 5'UTR, whereas the latter displayed significantly higher oncolytic activity. Overall, we presented here valid strategies to enhance the genomic stability of miR-CVB3 for virotherapy.
Collapse
Affiliation(s)
- Huitao Liu
- Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada,Department of Experimental Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada,Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada,Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Yuan Chao Xue
- Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada,Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | | | - Honglin Luo
- Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada,Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada,Corresponding author Honglin Luo, Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada.
| |
Collapse
|
31
|
Abdel Rhman M, Owira P. The role of microRNAs in the pathophysiology, diagnosis, and treatment of diabetic cardiomyopathy. J Pharm Pharmacol 2022; 74:1663-1676. [PMID: 36130185 DOI: 10.1093/jpp/rgac066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022]
Abstract
INTRODUCTION Diabetic cardiomyopathy (DCM) is an end-point macrovascular complication associated with increased morbidity and mortality in 12% of diabetic patients. MicroRNAs (miRNAs) are small noncoding RNAs that can act as cardioprotective or cardiotoxic agents in DCM. METHODS We used PubMed as a search engine to collect and analyse data in published articles on the role of miRNAs on the pathophysiology, diagnosis and treatment of DCM. RESULTS MiRNAs play an essential role in the pathophysiology, diagnosis and treatment of DCM due to their distinct gene expression patterns in diabetic patients compared to healthy individuals. Advances in gene therapy have led to the discovery of potential circulating miRNAs, which can be used as biomarkers for DCM diagnosis and prognosis. Furthermore, targeted miRNA therapies in preclinical and clinical studies, such as using miRNA mimics and anti-miRNAs, have yielded promising results. Application of miRNA mimics and anti-miRNAs via different nanodrug delivery systems alleviate hypertrophy, fibrosis, oxidative stress and apoptosis of cardiomyocytes. CONCLUSION MiRNAs serve as attractive potential targets for DCM diagnosis, prognosis and treatment due to their distinctive expression profile in DCM development.
Collapse
Affiliation(s)
- Mahasin Abdel Rhman
- Department of Pharmacology, Discipline of Pharmaceutical Sciences, Molecular and Clinical Pharmacology Research Laboratory, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Peter Owira
- Department of Pharmacology, Discipline of Pharmaceutical Sciences, Molecular and Clinical Pharmacology Research Laboratory, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| |
Collapse
|
32
|
miR-133a-A Potential Target for Improving Cardiac Mitochondrial Health and Regeneration After Injury. J Cardiovasc Pharmacol 2022; 80:187-193. [PMID: 35500168 DOI: 10.1097/fjc.0000000000001279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/01/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT The various roles of muscle secretory factors and myokines have been well studied, but in recent decades, the role of myocyte-specific microRNAs (myomiRs) has gained momentum. These myomiRs are known to play regulatory roles in muscle health in general, both skeletal muscle and cardiac muscle. In this review, we have focused on the significance of a myomiR termed miR-133a in cardiovascular health. The available literature supports the claim that miR-133a could be helpful in the healing process of muscle tissue after injury. The protective function could be due to its regulatory effect on muscle or stem cell mitochondrial function. In this review, we have shed light on the protective mechanisms offered by miR-133a. Most of the beneficial effects are due to the presence of miR-133a in circulation or tissue-specific expression. We have also reviewed the potential mechanisms by which miR-133a could interact with cell surface receptors and also transcriptional mechanisms by which they offer cardioprotection and regeneration. Understanding these mechanisms will help in finding an ideal strategy to repair cardiac tissue after injury.
Collapse
|
33
|
Abstract
Diabetes is a metabolic disorder that affects millions of people worldwide. Diabetic heart disease (DHD) comprises coronary artery disease, heart failure, cardiac autonomic neuropathy, peripheral arterial disease, and diabetic cardiomyopathy. The onset and progression of DHD have been attributed to molecular alterations in response to hyperglycemia in diabetes. In this context, microRNAs (miRNAs) have been demonstrated to have a significant role in the development and progression of DHD. In addition to their effects on the host cells, miRNAs can be released into circulation after encapsulation within the exosomes. Exosomes are extracellular nanovesicles ranging from 30 to 180 nm in diameter secreted by all cell types. They carry diverse cargos that are altered in response to various conditions in their parent cells. Exosomal miRNAs have been extensively studied in recent years due to their role and therapeutic potential in DHD. This review will first provide an overview of exosomes, their biogenesis and function, followed by the role of exosomes in cardiovascular disease and then focuses on the known role of exosomes and associated miRNAs in DHD.
Collapse
Affiliation(s)
- Dhananjie Chandrasekera
- Department of Physiology, School of Biomedical Sciences, HeartOtago, University of Otago, 270, Great King Street, Dunedin, New Zealand.
| | - Rajesh Katare
- Department of Physiology, School of Biomedical Sciences, HeartOtago, University of Otago, 270, Great King Street, Dunedin, New Zealand.
| |
Collapse
|
34
|
Seferović P, Farmakis D, Bayes-Genis A, Ben Gal T, Böhm M, Chioncel O, Ferrari R, Filippatos G, Hill L, Jankowska E, Lainscak M, Lopatin Y, Lund LH, Mebazaa A, Metra M, Moura B, Rosano G, Thum T, Voors A, Coats AJS. Biomarkers for the prediction of heart failure and cardiovascular events in patients with type 2 diabetes: a position statement from the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2022; 24:1162-1170. [PMID: 35703329 DOI: 10.1002/ejhf.2575] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/21/2022] [Accepted: 06/13/2022] [Indexed: 11/08/2022] Open
Abstract
Knowledge on risk predictors of incident heart failure (HF) in patients with type 2 diabetes (T2D) is crucial given the frequent coexistence of the two conditions and the fact that T2D doubles the risk of incident HF. In addition, HF is increasingly being recognized as an important endpoint in trials in T2D. On the other hand, the diagnostic and prognostic performance of established cardiovascular biomarkers may be modified by the presence of T2D. The present position paper, derived by an expert panel workshop organized by the Heart Failure Association of the European Society of Cardiology, summarizes the current knowledge and gaps in evidence regarding the use of a series of different biomarkers, reflecting various pathogenic pathways, for the prediction of incident HF and cardiovascular events in patients with T2D and in those with established HF and T2D.
Collapse
Affiliation(s)
- Peter Seferović
- Faculty of Medicine, University of Belgrade Belgrade, Serbia and Serbian Academy of Sciences and Arts, Belgrade, Serbia.,University of Belgrade Belgrade, Belgrade, Serbia
| | | | - Antoni Bayes-Genis
- Heart Institute, Hospital Universitari German Trias i Pujol, Badalona, Spain.,Department of Medicine, Universitat Autónoma de Barcelona, Barcelona, Spain.,CIBERCV, Instituto de Salud, Madrid, Spain
| | - Tuvia Ben Gal
- Heart Failure Unit, Cardiology Department, Rabin Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael Böhm
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Saarland University, Homburg, Germany
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases 'Prof. C.C. Iliescu', Bucharest, and University of Medicine Carol Davila, Bucharest, Romania
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Gerasimos Filippatos
- Second Department of Cardiology, Athens University Hospital Attikon, National and Kapodistrina University of Athens Medical School, Athens, Greece
| | - Loreena Hill
- School of Nursing and Midwifery, Queen's University, Belfast, UK
| | - Ewa Jankowska
- Department of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland.,Centre for Heart Diseases, University Hospital, Wroclaw, Poland
| | - Mitja Lainscak
- Division of Cardiology, General Hospital Murska Sobota, Murska Sobota, Slovenia, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Yuri Lopatin
- Volgograd State Medical University, Regional Cardiology Centre Volgograd, Volgograd, Russian Federation
| | - Lars H Lund
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Alexandre Mebazaa
- INSERM UMR-S 942, Paris, France; Department of Anesthesiology and Critical Care Medicine, St. Louis and Lariboisère University Hospitals, Paris, France
| | - Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | - Brenda Moura
- CINTESIS - Centro de Investigação em Tecnologias e Serviços de Saúde, Porto, Portugal; Serviço de Cardiologia, Hospital das Forças Armadas - Pólo do Porto, Porto, Portugal
| | - Giuseppe Rosano
- Cardiovascular Clinical Academic Group, St George's Hospitals NHS Trust University of London, London, UK.,IRCCS San Raffaele Pisana, Rome, Italy
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Adriaan Voors
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | |
Collapse
|
35
|
Dietary Acid Load Modulation of Asthma-Related miRNAs in the Exhaled Breath Condensate of Children. Nutrients 2022; 14:nu14061147. [PMID: 35334803 PMCID: PMC8949211 DOI: 10.3390/nu14061147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/22/2022] [Accepted: 03/04/2022] [Indexed: 01/27/2023] Open
Abstract
Individual nutrients and bioactive compounds have been implicated in the expression of microRNAs (miRNAs), which are related to inflammation and asthma. However, evidence about the impact of diet is scarce. Therefore, we aimed to assess the association between dietary acid load and asthma-related miRNA in the exhaled breath condensate (EBC) of school-aged children. This cross-sectional analysis included 150 participants aged 7 to 12 years (52% girls) from a nested case–control study, which randomly selected 186 children attending 71 classrooms from 20 public schools located in city of Porto, Portugal. Dietary data were collected by one 24 h-recall questionnaire. Dietary acid load was assessed using the potential renal acid load (PRAL) and net endogenous acid production (NEAP) scores. Based on previous studies, eleven asthma-related miRNAs were chosen and analyzed in EBC by reverse transcription-quantitative real-time PCR. PRAL, NEAP and miRNAs were categorized as high or low according to the median. Logistic regression models were performed to assess the association between dietary acid load scores and miRNAs. Children in high dietary acid load groups (PRAL ≥ 14.43 and NEAP ≥ 55.79 mEq/day) have significantly increased odds of having high miR-133a-3p levels. In conclusion, higher dietary acid loads possibly modulate asthma-related miRNAs of school-aged children.
Collapse
|
36
|
De Sousa RAL, Improta-Caria AC. Regulation of microRNAs in Alzheimer´s disease, type 2 diabetes, and aerobic exercise training. Metab Brain Dis 2022; 37:559-580. [PMID: 35075500 DOI: 10.1007/s11011-022-00903-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia. The evolution and aggregation of amyloid beta (β) oligomers is linked to insulin resistance in AD, which is also the major characteristic of type 2 diabetes (T2D). Being physically inactive can contribute to the development of AD and/or T2D. Aerobic exercise training (AET), a type of physical exercise, can be useful in preventing or treating the negative outcomes of AD and T2D. AD, T2D and AET can regulate the expression of microRNAs (miRNAs). Here, we review some of the changes in miRNAs expression regulated by AET, AD and T2D. MiRNAs play an important role in the gene regulation of key signaling pathways in both pathologies, AD and T2D. MiRNA dysregulation is evident in AD and has been associated with several neuropathological alterations, such as the development of a reactive gliosis. Expression of miRNAs are associated with many pathophysiological mechanisms involved in T2D like insulin synthesis, insulin resistance, glucose intolerance, hyperglycemia, intracellular signaling, and lipid profile. AET regulates miRNAs levels. We identified 5 miRNAs (miR-21, miR-29a/b, miR-103, miR-107, and miR-195) that regulate gene expression and are modulated by AET on AD and T2D. The identified miRNAs are potential targets to treat the symptoms of AD and T2D. Thus, AET is a non-pharmacological tool that can be used to prevent and fight the negative outcomes in AD and T2D.
Collapse
Affiliation(s)
- Ricardo Augusto Leoni De Sousa
- Programa Multicêntrico de Pós-Graduação Em Ciências Fisiológicas- Sociedade Brasileira de Fisiologia (SBFis), Universidade Federal Dos Vales Do Jequitinhonha E Mucuri (UFVJM), Campus JK, Rodovia MGT 367, Km 583, Alto da Jacuba, nº 5000, Diamantina, Minas Gerais, CEP 39100-000, Brazil.
| | - Alex Cleber Improta-Caria
- Post-Graduate Program in Medicine and Health, Faculty of Medicine, Federal University of Bahia, Bahia, Brazil
| |
Collapse
|
37
|
Yin Z, Chen C. Biological Functions and Clinical Prospects of Extracellular Non-Coding RNAs in Diabetic Cardiomyopathy: an Updated Review. J Cardiovasc Transl Res 2022; 15:469-476. [PMID: 35175553 DOI: 10.1007/s12265-022-10217-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/09/2022] [Indexed: 12/17/2022]
Abstract
Diabetic cardiomyopathy (DCM) is one of the major causes of heart failure in diabetic patients. However, the pathogenesis of diabetic cardiomyopathy has not been fully elucidated. Diagnosis and therapeutic strategy of DCM is still challenging. Various non-coding RNAs (ncRNA) are implicated in the onset and progression of DCM. Interestingly, ncRNAs not only are regulators intracellularly, but also can exist and function in extracellular space. Recent evidences have demonstrated that extracellular ncRNAs play emerging roles in both intracardiac and inter-organ communication during the pathogenesis of DCM; thus, extracellular ncRNAs are attractive diagnostic biomarkers and potential therapeutic targets for DCM. This article will review the current knowledge of the roles of extracellular ncRNAs in DCM, especially focusing on their physio-pathological properties and perspectives of potential clinical translation for biomarkers and therapies. Recent evidences have demonstrated that extracellular ncRNA play emerging roles in both intracardiac and inter-organ communication involved in the pathogenesis of diabetic cardiomyopathy (DCM), thus shown as attractive diagnostic biomarkers and potential therapeutics for DCM. In the current review, we first summarize the progress regarding the paracrine role of extracellular ncRNA in DCM. miRNAs and circRNAs have been shown to mediate the communication among cardiomyocytes, endothelial cells, and vascular smooth muscle cells in the diabetic heart. Subsequently, we systematically describe that extracellular ncRNAs contribute to the crosstalk between the heart and other organs in the context of diabetes. Researches have indicated that miRNAs acted as hepatokines and adipokines to mediates the injure effect of distal organs on hearts. As for clinical application, extracellular ncRNAs are promising biomarker and have therapeutic potential. (Created with BioRender.com).
Collapse
Affiliation(s)
- Zhongwei Yin
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China.
| |
Collapse
|
38
|
Rodríguez-Sanabria JS, Escutia-Gutiérrez R, Rosas-Campos R, Armendáriz-Borunda JS, Sandoval-Rodríguez A. An Update in Epigenetics in Metabolic-Associated Fatty Liver Disease. Front Med (Lausanne) 2022; 8:770504. [PMID: 35087844 PMCID: PMC8787199 DOI: 10.3389/fmed.2021.770504] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is characterized by hepatic steatosis accompanied by one of three features: overweight or obesity, T2DM, or lean or normal weight with evidence of metabolic dysregulation. It is distinguished by excessive fat accumulation in hepatocytes, and a decrease in the liver's ability to oxidize fats, the accumulation of ectopic fat, and the activation of proinflammatory pathways. Chronic damage will keep this pathophysiologic cycle active causing progression from hepatic steatosis to cirrhosis and eventually, hepatocarcinoma. Epigenetics affecting gene expression without altering DNA sequence allows us to study MAFLD pathophysiology from a different perspective, in which DNA methylation processes, histone modifications, and miRNAs expression have been closely associated with MAFLD progression. However, these considerations also faced us with the circumstance that modifying those epigenetics patterns might lead to MAFLD regression. Currently, epigenetics is an area of great interest because it could provide new insights in therapeutic targets and non-invasive biomarkers. This review comprises an update on the role of epigenetic patterns, as well as innovative therapeutic targets and biomarkers in MAFLD.
Collapse
Affiliation(s)
- J Samael Rodríguez-Sanabria
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara, Mexico
| | - Rebeca Escutia-Gutiérrez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara, Mexico
| | - Rebeca Rosas-Campos
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara, Mexico
| | - Juan S Armendáriz-Borunda
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara, Mexico.,School of Medicine and Health Sciences, Tecnologico de Monterrey, Campus Guadalajara, Zapopan, Mexico
| | - Ana Sandoval-Rodríguez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
39
|
Improta-Caria AC, De Sousa RAL, Roever L, Fernandes T, Oliveira EMD, Aras Júnior R, Souza BSDF. MicroRNAs in type 2 diabetes mellitus: potential role of physical exercise. Rev Cardiovasc Med 2022; 23:29. [PMID: 35092221 DOI: 10.31083/j.rcm2301029] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/19/2021] [Accepted: 11/25/2021] [Indexed: 08/08/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a multifactorial metabolic disease, and its prevalence has grown worldwide. Several pathophysiological processes contribute to the development, progression and aggravating of the disease, for example, decreased insulin synthesis and secretion, insulin resistance, inflammation, and apoptosis, all these processes are regulated by various epigenetic factors, including microRNAs (miRNAs). MiRNAs are small non-coding RNAs, which are around 20 nucleotides in length and are regulators of gene expression at the post-transcriptional level, have a specific function of inhibiting or degrading a messenger RNA target. Thus, miRNAs modulate the expression of many associated genes with the pathophysiological processes in T2DM. On the other hand, miRNAs are also modulated through physical exercise (PE), which induces a change in their expression pattern during and after exercise. Some scientific evidence shows that PE modulates miRNAs beneficially and improves the signaling pathway of insulin resistance, however, little is known about the function of PE modulating miRNAs associated with the processes of insulin secretion, inflammation, and apoptosis. Thus, the objective of this review is to identify the miRNAs expression pattern in T2DM and compare it with the exercise-induced miRNAs expression pattern, identifying the signaling pathways that these miRNAs are regulating in the processes of insulin secretion, insulin resistance, inflammation, and apoptosis in T2DM, and how PE may have a potential role in modulating these signal transduction pathways, promoting benefits for patients with T2DM.
Collapse
Affiliation(s)
- Alex Cleber Improta-Caria
- Post-Graduate Program in Medicine and Health, Faculty of Medicine, Federal University of Bahia, 40110-100 Bahia, Brazil
- Center for Biotechnology and Cell Therapy, Sao Rafael Hospital, 41253-190 Bahia, Brazil
- Department of Physical Education in Cardiology, Brazilian Society of Cardiology, 41170-130 Bahia, Brazil
| | - Ricardo Augusto Leoni De Sousa
- Physiological Science Multicentric Program, Federal University of Valleys' Jequitinhonha and Mucuri, 39960-000 Minas Gerais, Brazil
| | - Leonardo Roever
- Federal University of Uberlandia, 38400-128 Minas Gerais, Brazil
| | - Tiago Fernandes
- Laboratory of Biochemistry and Molecular Biology of the Exercise, School of Physical Education and Sport, Sao Paulo University, 05508-030 Sao Paulo, Brazil
| | - Edilamar Menezes de Oliveira
- Laboratory of Biochemistry and Molecular Biology of the Exercise, School of Physical Education and Sport, Sao Paulo University, 05508-030 Sao Paulo, Brazil
| | - Roque Aras Júnior
- Post-Graduate Program in Medicine and Health, Faculty of Medicine, Federal University of Bahia, 40110-100 Bahia, Brazil
| | - Bruno Solano de Freitas Souza
- Center for Biotechnology and Cell Therapy, Sao Rafael Hospital, 41253-190 Bahia, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), 40296-710 Bahia, Brazil
- D'Or Institute for Research and Education (IDOR), 41253-190 Salvador, Brazil
| |
Collapse
|
40
|
Meza CA, Amador M, McAinch AJ, Begum K, Roy S, Bajpeyi S. Eight weeks of combined exercise training do not alter circulating microRNAs-29a, -133a, -133b, and -155 in young, healthy men. Eur J Appl Physiol 2022; 122:921-933. [PMID: 35015112 DOI: 10.1007/s00421-022-04886-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/04/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Individuals with a family history of type 2 diabetes (FH +) have an increased risk of developing type 2 diabetes. Circulating microRNAs (miRNAs) have been implicated as biomarkers of type 2 diabetes risk. Here, we investigated if four circulating miRNAs related to glucose metabolism were altered in men with a FH + and we conducted a preliminary analysis to determine if miRNA expressions were responsive to 8 weeks of combined exercise training. METHODS Sixteen young healthy men (mean ± SD; age 22.5 ± 2.5; BMI 26.4 ± 4.0) with FH + or without a family history of type 2 diabetes (FH -) underweight 8 weeks of combined endurance and resistance exercise training (n = 8 FH -; n = 8 FH +). The expression of miR-29a, miR-133a, miR-133b, and miR-155 were measured in serum before and after exercise training. QIAGEN's Ingenuity® Pathway Analysis was used to examine miRNA target genes and their involvement in glucose metabolism signaling pathways. RESULTS There were no differences in miRNA expressions between FH - and FH + . Exercise training did not alter miRNA expressions in either FH - or FH + despite improvements in insulin sensitivity, aerobic capacity, and muscular strength. miR-29a and miR-155 were inversely related to fasting glucose, and miR-133a and miR-133b were negatively correlated with glucose tolerance; however, correlations were not observed with insulin sensitivity. CONCLUSIONS The circulating miRNAs- miR-29a, miR-133a, miR-133b, and miR-155 are related to measures of glucose metabolism in healthy, normoglycemic men, but do not reflect peripheral insulin sensitivity or improvements in metabolic health following 8 weeks of combined exercise training.
Collapse
Affiliation(s)
- Cesar A Meza
- Metabolic, Nutrition and Exercise Research (MiNER) Laboratory, Department of Kinesiology, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79902, USA
| | - Manuel Amador
- Metabolic, Nutrition and Exercise Research (MiNER) Laboratory, Department of Kinesiology, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79902, USA
| | - Andrew J McAinch
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, VIC, Australia
| | - Khodeza Begum
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79902, USA
| | - Sourav Roy
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79902, USA
| | - Sudip Bajpeyi
- Metabolic, Nutrition and Exercise Research (MiNER) Laboratory, Department of Kinesiology, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79902, USA.
| |
Collapse
|
41
|
Preethi KA, Selvakumar SC, Sekar D. Diagnostic and Therapeutic Application of Exosomal microRNAs Inducing Inflammation in Type 2 Diabetes Mellitus. Crit Rev Immunol 2022; 42:1-11. [PMID: 36374817 DOI: 10.1615/critrevimmunol.2022044927] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Diabetes mellitus is a class of noncommunicable chronic metabolic disorders marked by hyperglycemia due to insulin production, insulin action or both and has reached epidemic levels around the world. The two most frequent types of diabetes are type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). Despite substantial improvements in the knowledge and treatment of DM, the associated incidence and mortality rates remain steadily increased. Reliable markers for the early detection, monitoring and focused treatment of DM are desperately required. Conversely, microRNAs (miRNAs) have received much significance due to their regulatory involvement in gene expression. Fascinatingly, exosomes can be enclosed into miRNAs to transport or distribute them into the target cells or tissues in which they have a physiological regulatory action. Thus, exosomal miRNAs are proving to be important regulators in the establishment and maintenance of DM, however, further mode of action will be needed to investigate in order to fully comprehend the pathophysiological process. Hereby, this review outlines the recent findings on the role of exosomal miRNAs intending to understand the precise function in diagnostic and therapeutic aspects in T2DM disease.
Collapse
Affiliation(s)
- K Auxzilia Preethi
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, India
| | - Sushmaa Chandralekha Selvakumar
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, India
| | - Durairaj Sekar
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, India
| |
Collapse
|
42
|
Li D, Wang P, Wei W, Wang C, Zhong Y, Lv L, Wang J. Serum MicroRNA Expression Patterns in Subjects After the 5-km Exercise Are Strongly Associated With Cardiovascular Adaptation. Front Physiol 2021; 12:755656. [PMID: 34912238 PMCID: PMC8667031 DOI: 10.3389/fphys.2021.755656] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/01/2021] [Indexed: 12/02/2022] Open
Abstract
Circulating microRNAs (miRNAs) have been reported dysregulated during exercise. However, the changes of specific serum miRNAs during the 5-km run test with intensity of 51–52% maximum oxygen uptake (V̇O2max) and their association with traditional cardiovascular-related indicators remain well-characterized. Levels of miR-1, miR-21, miR-146a, miR-155, miR-181, and miR-210 were detected in 120 young subjects before and after the exercise training by quantitative reverse-transcription PCR (RT-qPCR). Besides, the levels of cardiac troponin I (cTNI), myoglobin (Myo), creatine kinase (CK), creatine kinase-MB (CK-MB), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), ischemia-modified albumin (IMA), interleukin-6 (IL-6), and C-reactive protein (CRP) were measured and the correlation between levels of serum miRNAs and biochemical parameters was also analyzed. Compared with resting state, the serum levels of miR-1, miR-146a, miR-155, miR-181, and miR-210 were significantly increased after exercise training. Serum levels of miR-146a, miR-155, and miR-210 after exercise training were positively correlated with Myo, CK-MB, and LDH, respectively, while miR-1, miR-146a, miR-181, and miR-155 were positively correlated with the levels of IL-6. Additionally, all the five miRNAs were negatively correlated with IMA levels. The multivariate logistic regression analysis showed that high levels of miR-146a, AST, LDH, and IL-6 in serum were risk factors, while low IMA contents were a protective factor for cardiovascular adaptation during exercise. In conclusion, the dynamic changes of miRNAs under the condition of the 5-km continuous running contribute to the adaptive regulation of the cardiovascular function of the body.
Collapse
Affiliation(s)
- Dandan Li
- Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Pingping Wang
- Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Wenyan Wei
- Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Cheng Wang
- Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yong Zhong
- Department of Health Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Lei Lv
- Department of Geriatric Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Junjun Wang
- Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
43
|
Cione E, Cannataro R, Gallelli L, De Sarro G, Caroleo MC. Exosome microRNAs in Metabolic Syndrome as Tools for the Early Monitoring of Diabetes and Possible Therapeutic Options. Pharmaceuticals (Basel) 2021; 14:1257. [PMID: 34959658 PMCID: PMC8706321 DOI: 10.3390/ph14121257] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes are nano-sized extracellular vesicles produced and released by almost all cell types. They play an essential role in cell-cell communications by delivering cellular bioactive compounds such as functional proteins, metabolites, and nucleic acids, including microRNA, to recipient cells. Thus, they are involved in various physio-pathological conditions. Exosome-miRNAs are associated with numerous diseases, including type 2 diabetes, a complex multifactorial metabolic disorder linked to obesity. In addition, exosome-miRNAs are emerging as essential regulators in the progression of diabetes, principally for pancreatic β-cell injury and insulin resistance. Here, we have clustered the recent findings concerning exosome-miRNAs associated with β-cell dysfunction to provide a novel approach for the early diagnosis and therapy of diabetes.
Collapse
Affiliation(s)
- Erika Cione
- Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, Ed. Polifunzionale, Arcavacata di Rende, 87036 Rende, CS, Italy;
- GalaScreen Laboratories, University of Calabria, Ed. Polifunzionale, Arcavacata di Rende, 87036 Rende, CS, Italy;
| | - Roberto Cannataro
- GalaScreen Laboratories, University of Calabria, Ed. Polifunzionale, Arcavacata di Rende, 87036 Rende, CS, Italy;
| | - Luca Gallelli
- Department of Health Science, University of Catanzaro and Operative Unit of Clinical Pharmacology and Pharmacovigilance, Mater Domini Hospital, 88100 Catanzaro, CZ, Italy; (L.G.); (G.D.S.)
| | - Giovambattista De Sarro
- Department of Health Science, University of Catanzaro and Operative Unit of Clinical Pharmacology and Pharmacovigilance, Mater Domini Hospital, 88100 Catanzaro, CZ, Italy; (L.G.); (G.D.S.)
| | - Maria Cristina Caroleo
- Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, Ed. Polifunzionale, Arcavacata di Rende, 87036 Rende, CS, Italy;
- GalaScreen Laboratories, University of Calabria, Ed. Polifunzionale, Arcavacata di Rende, 87036 Rende, CS, Italy;
| |
Collapse
|
44
|
Sałówka A, Martinez-Sanchez A. Molecular Mechanisms of Nutrient-Mediated Regulation of MicroRNAs in Pancreatic β-cells. Front Endocrinol (Lausanne) 2021; 12:704824. [PMID: 34803905 PMCID: PMC8600252 DOI: 10.3389/fendo.2021.704824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic β-cells within the islets of Langerhans respond to rising blood glucose levels by secreting insulin that stimulates glucose uptake by peripheral tissues to maintain whole body energy homeostasis. To different extents, failure of β-cell function and/or β-cell loss contribute to the development of Type 1 and Type 2 diabetes. Chronically elevated glycaemia and high circulating free fatty acids, as often seen in obese diabetics, accelerate β-cell failure and the development of the disease. MiRNAs are essential for endocrine development and for mature pancreatic β-cell function and are dysregulated in diabetes. In this review, we summarize the different molecular mechanisms that control miRNA expression and function, including transcription, stability, posttranscriptional modifications, and interaction with RNA binding proteins and other non-coding RNAs. We also discuss which of these mechanisms are responsible for the nutrient-mediated regulation of the activity of β-cell miRNAs and identify some of the more important knowledge gaps in the field.
Collapse
Affiliation(s)
| | - Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
45
|
Jin ZQ. MicroRNA targets and biomarker validation for diabetes-associated cardiac fibrosis. Pharmacol Res 2021; 174:105941. [PMID: 34656765 DOI: 10.1016/j.phrs.2021.105941] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/06/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022]
Abstract
Cardiac fibrosis is one of the main characteristics of diabetic cardiomyopathy and manifests excessive accumulation of extracellular matrix proteins in the heart. Several signaling pathways have been proposed for pathogenesis of cardiac fibrosis in the diabetic heart. TGF-β/Smad2/3-dependent or independent pathway is the major signaling molecule core in the pathogenesis of cardiac fibrosis. MicroRNAs (miRNAs, miR) are ~22-nuceotide regulatory RNAs that are involved in gene silencing through the degradation of post-transcriptional mRNA or suppression of the expressed proteins. Hyperglycemia in the diabetic heart regulates expression of some miRNAs. Target molecules of miRNAs can be identified through biocomputational database initial screening and dual luciferase assay validation. miR-21, miR-150-5p, miR-155, miR-216a-3p, miR-221-3p, miR-223, and miR-451 were up-regulated in the diabetic heart and promoted cardiac fibrosis through targeting signaling pathways in cardiac fibroblasts, endothelial cells, and cardiac myocytes. miR-15a/-15b, miR-18a-5p, miR-20a-5p, miR-26b-5p, miR-29, miR-133a, miR-141, miR-146, miR-200b, miR-203, miR-222, and miR-551b-5p were down-regulated in the diabetic heart and exhibited anti-fibrosis when they were overexpressed. miRNAs are stable molecules and may reflect the pathological changes of organs. Some miRNAs have been detected in the plasma or serum in patients with diabetes mellitus or heart failure. Exploration of targets and biomarkers of miRNA may provide additional information on pathogenesis and diagnosis of cardiac fibrosis and novel targets to tackle diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Zhu-Qiu Jin
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, CA 95757, USA.
| |
Collapse
|
46
|
Li X, Li N, Li B, Feng Y, Zhou D, Chen G. Noncoding RNAs and RNA-binding proteins in diabetic wound healing. Bioorg Med Chem Lett 2021; 50:128311. [PMID: 34438011 DOI: 10.1016/j.bmcl.2021.128311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Poor wound healing is a common complication in diabetic patients. It often leads to intractable infections and lower limb amputations and is associated with cardiovascular morbidity and mortality. NcRNAs, which can regulate gene expression, have emerged as important regulators of various physiological processes. Herein, we summarize the diverse roles of ncRNAs in the key stages of diabetic wound healing, including inflammation, angiogenesis, re-epithelialization, and extracellular matrix remodeling. Meanwhile, the potential use of ncRNAs as novel therapeutic targets for wound healing in diabetic patients is also discussed. In addition, we summarize the role of RNA-binding proteins (RBPs) in the regulation of gene expression and signaling pathways during skin repair, which may provide opportunities for therapeutic intervention for this potentially devastating disease. However, so far, research on the modulated drug based on ncRNAs that lead to significantly altered gene expression in diabetic patients is scarce. We have compiled some drugs that may be able to modulate ncRNAs, which significantly regulate the gene expression in diabetic patients.
Collapse
Affiliation(s)
- Xue Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Ning Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Bingxin Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yuan Feng
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Di Zhou
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Gang Chen
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China; Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, People's Republic of China.
| |
Collapse
|
47
|
He XY, Ou CL. Clinical significance of serum miR-129-5p in patients with diabetes mellitus presenting macrovascular complications. World J Diabetes 2021; 12:1282-1291. [PMID: 34512893 PMCID: PMC8394230 DOI: 10.4239/wjd.v12.i8.1282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/24/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diabetic macrovascular complications (DMCs) are the most common complications encountered during the course of diabetes mellitus (DM) with extremely high mortality rates. Therefore, there is an urgent need to identify specific and sensitive biomarkers for the early diagnosis of DMCs. AIM To investigate the expression and significance of serum miR-129-5p in patients with DM and macrovascular complications. METHODS Serum samples were collected from 36 healthy controls, 58 patients with DM presenting no macrovascular complications, and 62 patients with DMCs. The expression of miR-129-5p was detected using quantitative real-time polymerase chain reaction. Pearson's correlation assay was performed to analyze the correlation between serum miR-129-5p levels and clinical indicators. Receiver operator characteristic (ROC) analysis was conducted to analyze the diagnostic value of serum miR-129-5p in patients with DM or DMCs. RESULTS There was a 4.378-fold and 7.369-fold increase in serum miR-129-5p expression in the DM (5.346 ± 0.405) and DMCs (8.998 ± 0.631) groups, respectively (P < 0.001), compared with the control group (1.221±0.090). In addition, the expression of serum miR-129-5p in patients with DMCs was higher than that in patients with DM, revealing a 1.683-fold increase (P < 0.001). Additionally, serum miR-129-5p expression significantly correlated with smoking history, disease duration, and glycated hemoglobin (HbA1c) in patients with DMCs (P < 0.001). The area under the ROC curve (AUC) of miR-129-5p as a serum marker was 0.964 (95% confidence interval [CI]: 0.930-0.997, P < 0.001) in distinguishing between patients with DM and healthy controls, whereas the AUC of miR-129-5p as a serum marker was 0.979 (95%CI: 0.959-0.999, P < 0.001) in distinguishing between patients with DMCs and healthy controls. CONCLUSION Elevated serum miR-129-5p expression levels correlate with the development of DMCs and can be utilized as a novel early diagnostic biomarker for DM combined with macrovascular complications.
Collapse
Affiliation(s)
- Xiao-Yun He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Chun-Lin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
48
|
Salvatore T, Pafundi PC, Galiero R, Albanese G, Di Martino A, Caturano A, Vetrano E, Rinaldi L, Sasso FC. The Diabetic Cardiomyopathy: The Contributing Pathophysiological Mechanisms. Front Med (Lausanne) 2021; 8:695792. [PMID: 34277669 PMCID: PMC8279779 DOI: 10.3389/fmed.2021.695792] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Individuals with diabetes mellitus (DM) disclose a higher incidence and a poorer prognosis of heart failure (HF) than non-diabetic people, even in the absence of other HF risk factors. The adverse impact of diabetes on HF likely reflects an underlying “diabetic cardiomyopathy” (DM–CMP), which may by exacerbated by left ventricular hypertrophy and coronary artery disease (CAD). The pathogenesis of DM-CMP has been a hot topic of research since its first description and is still under active investigation, as a complex interplay among multiple mechanisms may play a role at systemic, myocardial, and cellular/molecular levels. Among these, metabolic abnormalities such as lipotoxicity and glucotoxicity, mitochondrial damage and dysfunction, oxidative stress, abnormal calcium signaling, inflammation, epigenetic factors, and others. These disturbances predispose the diabetic heart to extracellular remodeling and hypertrophy, thus leading to left ventricular diastolic and systolic dysfunction. This Review aims to outline the major pathophysiological changes and the underlying mechanisms leading to myocardial remodeling and cardiac functional derangement in DM-CMP.
Collapse
Affiliation(s)
- Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gaetana Albanese
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Anna Di Martino
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
49
|
Lu YK, Chu X, Wang S, Sun Y, Zhang J, Dong J, Yan YX. Identification of Circulating hsa_circ_0063425 and hsa_circ_0056891 as Novel Biomarkers for Detection of Type 2 Diabetes. J Clin Endocrinol Metab 2021; 106:e2688-e2699. [PMID: 33608702 DOI: 10.1210/clinem/dgab101] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/17/2022]
Abstract
CONTEXT Circular RNAs (circRNAs), which are involved in the development of diseases by regulating gene expression, have become promising novel biomarkers for diseases. OBJECTIVE The aim of the present study was to identify the circulating circRNA biomarkers for early detection of type 2 diabetes (T2D). METHODS The circRNA expression profiles were screened by microarray and compared between 5 new T2D cases and 5 healthy controls. The expression of candidate circRNAs that may be involved in the insulin phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathway were validated by RT-qPCR in a second sample with 30 T2D cases and 30 controls. The association between circRNAs and T2D and their clinical significances were further assessed by logistic regression model, correlation analysis, and ROC curve in a large cohort comprising 313 subjects. The microRNA (miRNA) targets of circRNAs were verified by dual-luciferase reporter assay and RNA immunoprecipitation assay. RESULTS Low expressed circ_0063425 and hsa_circ_0056891 were independent predictors of T2D, impaired fasting glucose (IFG), and insulin resistance. The 2-circRNA panel had a high diagnostic accuracy for discriminating T2D and IFG from healthy controls, especially when body mass index was integrated. miR-19a-3p and miR-1-3p were identified as the miRNA targets of hsa_circ_0063425 and hsa_circ_0056891, respectively. Significant positive correlations were found between the expression levels of AKT and hsa_circ_0063425, PI3K and hsa_circ_0056891, in the total sample and subgroups stratified by glucose levels. CONCLUSION Downregulated hsa_circ_0063425 and hsa_circ_0056891 might contribute to the pathogenesis of T2D. They are valuable circulating biomarkers for early detection of T2D, which may be involved in regulation of PI3K/AKT signaling.
Collapse
Affiliation(s)
- Ya-Ke Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Xi Chu
- Health Management Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shuo Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Yue Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Jie Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Municipal Key Laboratory of Clinical Epidemiology, Beijing 100069, China
| | - Jing Dong
- Health Management Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yu-Xiang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Municipal Key Laboratory of Clinical Epidemiology, Beijing 100069, China
| |
Collapse
|
50
|
He X, Kuang G, Wu Y, Ou C. Emerging roles of exosomal miRNAs in diabetes mellitus. Clin Transl Med 2021; 11:e468. [PMID: 34185424 PMCID: PMC8236118 DOI: 10.1002/ctm2.468] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Exosomes are small extracellular vesicles 40-160 nm in diameter that are secreted by almost all cell types. Exosomes can carry diverse cargo including RNA, DNA, lipids, proteins, and metabolites. Exosomes transfer substances and information between cells by circulating in body fluids and are thus involved in diverse physiological and pathological processes in the human body. Recent studies have closely associated exosomal microRNAs (miRNAs) with various human diseases, including diabetes mellitus (DM), which is a complex multifactorial metabolic disorder disease. Exosomal miRNAs are emerging as pivotal regulators in the progression of DM, mainly in terms of pancreatic β-cell injury and insulin resistance. Exosomal miRNAs are closely associated with DM-associated complications, such as diabetic retinopathy (DR), diabetic nephropathy (DN), and diabetic cardiomyopathy (DCM), etc. Further investigations of the mechanisms of action of exosomal miRNAs and their role in DM will be valuable for the thorough understanding of the physiopathological process of DM. Here, we have summarized recent findings regarding exosomal miRNAs associated with DM to provide a new strategy for identifying potential diagnostic biomarkers and drug targets for the early diagnosis and treatment, respectively, of DM.
Collapse
Affiliation(s)
- Xiaoyun He
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Departments of Ultrasound Imaging, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Gaoyan Kuang
- Department of OrthopedicsThe First Affiliated Hospital of Hunan University of Chinese MedicineChangshaHunan410007China
- Postdoctoral Research WorkstationHinye Pharmaceutical Co. LtdChangshaHunan410331China
| | - Yongrong Wu
- Hunan university of Chinese MedicineChangshaHunan410208China
| | - Chunlin Ou
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| |
Collapse
|