1
|
Zhou H, Long Y, Yu F, Ji C, Gui L, Lu Y. Resveratrol improves gasdermin D-mediated pyroptosis of vascular endothelial cells induced by a high-fat diet and palmitic acid possibly via the SIRT1-p66Shc-NLRP3 pathway. J Nutr Biochem 2025; 140:109890. [PMID: 40054674 DOI: 10.1016/j.jnutbio.2025.109890] [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: 12/29/2023] [Revised: 01/15/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025]
Abstract
Resveratrol (RSV) ameliorates endothelial dysfunction (ED) primarily through sirtuin 1 (SIRT1). Increasing evidence shows pyroptosis as a novel mechanism in palmitic acid (PA)-induced ED. p66Shc is an adaptor protein involved in oxidative stress. However, whether RSV attenuates the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome via p66Shc remains elusive. This study aims to evaluate whether the antipyroptotic effect of RSV and the SIRT1 inhibitor EX527 are related to p66Shc. High-fat diet (HFD) induced obesity in mice, and RSV was administered intragastrically with 400mg/kg/d for 22 successive weeks. The serum levels of interleukin-1β (IL-1β) and IL-18 were analyzed, and the expression of related proteins were assayed with immunohistochemistry in the thoracic aorta. human umbilical vein endothelial cells (HUVECs) were induced by PA, then treated with RSV and EX527 respectively, lactate dehydrogenase (LDH) release, reactive oxygen species (ROS) levels, mitochondrial membrane potential (MMP) and expression of p66Shc, NLRP3, GSDMD and pyroptosis-related genes were assayed. RSV administration ameliorated endothelial cell pyroptosis by decreasing serum IL-1β and IL-18, the expression of NLRP3, p66Shc, and gasdermin D (GSDMD), and increasing the expression of SIRT1 in the HFD-treated thoracic aorta. PA promoted GSDMD-mediated endothelial cell pyroptosis by ROS production, LDH release, decreased MMP and SIRT1 expression, increased expression of p66Shc and activation of the NLRP3 inflammasome in a dose-dependent manner. RSV attenuated PA-induced pyroptosis, whereas EX527 reversed the antipyroptotic effect of RSV in PA-treated HUVECs. Our results suggested a new mechanism that RSV improves PA-induced pyroptosis in endothelial cells via the SIRT1-p66Shc-NLRP3 pathway.
Collapse
Affiliation(s)
- Heng Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yueming Long
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Guangdong Hydropower Group Hospital, Guangzhou, China
| | - Fangmei Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Chenhui Ji
- The Second Clinical Medical College of Anhui Medical University, Hefei, China
| | - Li Gui
- The Comprehensive Experimental Center, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yunxia Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China; The Comprehensive Experimental Center, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Zhang S, Liu J, Zhao H, Gao Y, Ren C, Zhang X. What do You Need to Know after Diabetes and before Diabetic Retinopathy? Aging Dis 2025:AD.2025.0289. [PMID: 40354381 DOI: 10.14336/ad.2025.0289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025] Open
Abstract
Diabetic retinopathy (DR) is a leading cause of vision impairment and blindness among individuals with diabetes mellitus. Current clinical diagnostic criteria mainly base on visible vascular structure changes, which are insufficient to identify diabetic patients without clinical DR (NDR) but with dysfunctional retinopathy. This review focuses on retinal endothelial cells (RECs), the first cells to sense and respond to elevated blood glucose. As blood glucose rises, RECs undergo compensatory and transitional phases, and the correspondingly altered molecules are likely to become biomarkers and targets for early prediction and treatment of NDR with dysfunctional retinopathy. This article elaborated the possible pathophysiological processes focusing on RECs and summarized recently published and reliable biomarkers for early screening and emerging intervention strategies for NDR patients with dysfunctional retinopathy. Additionally, references for clinical medication selection and lifestyle recommendations for this population are provided. This review aims to deepen the understanding of REC biology and NDR pathophysiology, emphasizes the importance of early detection and intervention, and points out future directions to improve the diagnosis and treatment of NDR with dysfunctional retinopathy and to reduce the occurrence of DR.
Collapse
Affiliation(s)
- Shiyu Zhang
- Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Laboratory for Clinical Medicine, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Heng Zhao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Laboratory for Clinical Medicine, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yuan Gao
- Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China
| | - Xuxiang Zhang
- Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Colasanti JJ, Lin JB, Terao R, Lee TJ, Santeford A, Apte RS. MicroRNA-34a suppresses KLF2 to promote pathological angiogenesis through the CXCR4/CXCL12 pathway in age-related macular degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637499. [PMID: 39990324 PMCID: PMC11844524 DOI: 10.1101/2025.02.12.637499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Age-related macular degeneration (AMD), characterized by pathologic choroidal neovascularization (CNV), is a leading cause of vision loss in the elderly. Vascular endothelial growth factor A (VEGFa) antagonists can prevent acute vision loss, but high treatment burden and loss of efficacy with chronic therapy highlight the need to explore alternative mechanisms. Recently, microRNA-34a (miR-34a) has emerged as a key regulator in aging and age-related diseases, but its role in neovascular AMD is unclear. In an injury-induced murine CNV model, we discovered miR-34a promoted pathological angiogenesis, without altering expression of Vegfa or its receptor Kdr, the canonical regulators of CNV. Mechanistically, miR-34a directly targets and inhibits the transcription factor KLF2 thereby upregulating the pro-angiogenic factors CXCR4 and CXCL12. Finally, we show miR-34a exacerbates CNV in aged mice and is expressed in CNV lesions excised from wet AMD patients. These findings establish a causal link between the age-related miR-34a and neovascularization in AMD. Teaser Identification of a molecular mechanism involved in the pathogenesis of a prevalent and debilitating age-related ocular disease.
Collapse
|
4
|
Iusupova AO, Pakhtusov NN, Slepova OA, Khabarova NV, Privalova EV, Bure IV, Nemtsova MV, Belenkov YN. MiRNA-34a, miRNA-145, and miRNA-222 Expression, Matrix Metalloproteinases, TNF-α and VEGF in Patients with Different Phenotypes of Coronary Artery Disease. Int J Mol Sci 2024; 25:12978. [PMID: 39684689 PMCID: PMC11641108 DOI: 10.3390/ijms252312978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
The development of different phenotypes of coronary artery (CA) lesions is regulated via many various factors, such as pro-inflammatory agents, zinc-dependent endopeptidases, growth factors and circulating microRNAs (miRs). To evaluate the expression levels of miR-34a, miR-145 and miR-222, tumor necrosis factor α (TNF-α), matrix metalloproteinases (MMP-1, -9, -13 and -14) and vascular endothelial growth factor (VEGF) in patients with different phenotypes of coronary artery disease (CAD): ischemia/angina with non-obstructive coronary arteries (INOCA/ANOCA) and obstructive CAD (oCAD) compared with a control group. This cross-sectional observational study included 157 subjects with a verified CAD diagnosis (51 patients with INOCA, 76 patients with oCAD and 30 healthy volunteers). The expression of miR-34a, miR-145 and miR-222 (RT-PCR) and the levels of VEGF, TNF-α, MMP-1, MMP-9, MMP-13 and MMP-14 (ELISA) were estimated in plasma samples. A higher concentration of MMP-9 was found in oCAD-group samples compared to the INOCA/ANOCA group. The INOCA/ANOCA group was characterized by higher levels of TNF-α. Based on multivariate regression analysis, a mathematical model predicting the type of CA lesion was constructed. MiR-145 was the independent predictor of INOCA/ANOCA (p = 0.006). Changes in concentrations of MMP-9 and MMP-14 were found in both investigated CAD groups, with MMP-9 levels being significantly higher in obstructive CAD samples than in INOCA/ANOCA, which confirms the role of inflammation in the development of atherosclerosis. A multivariate regression analysis allowed us to achieve a model that can predict the phenotype of stable CAD, and MiR-145 can be assumed as an independent predictor of INOCA/ANOCA.
Collapse
Affiliation(s)
- Alfiya Oskarovna Iusupova
- Department of Hospital Therapy No 1, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (N.N.P.); (O.A.S.); (N.V.K.); (E.V.P.); (Y.N.B.)
| | - Nikolay Nikolaevich Pakhtusov
- Department of Hospital Therapy No 1, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (N.N.P.); (O.A.S.); (N.V.K.); (E.V.P.); (Y.N.B.)
| | - Olga Alexandrovna Slepova
- Department of Hospital Therapy No 1, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (N.N.P.); (O.A.S.); (N.V.K.); (E.V.P.); (Y.N.B.)
| | - Natalia Vladimirovna Khabarova
- Department of Hospital Therapy No 1, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (N.N.P.); (O.A.S.); (N.V.K.); (E.V.P.); (Y.N.B.)
| | - Elena Vitalievna Privalova
- Department of Hospital Therapy No 1, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (N.N.P.); (O.A.S.); (N.V.K.); (E.V.P.); (Y.N.B.)
| | - Irina Vladimirovna Bure
- Laboratory of Medical Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (I.V.B.); (M.V.N.)
- Research Institute of Molecular and Personalized Medicine, Russian Medical Academy of Continuous Professional Education, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia
| | - Marina Vyacheslavovna Nemtsova
- Laboratory of Medical Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (I.V.B.); (M.V.N.)
- Laboratory of Epigenetics, Research Centre for Medical Genetics, 115522 Moscow, Russia
| | - Yuri Nikitich Belenkov
- Department of Hospital Therapy No 1, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (N.N.P.); (O.A.S.); (N.V.K.); (E.V.P.); (Y.N.B.)
| |
Collapse
|
5
|
Mohamed AA, Abdallah GM, Ibrahim IT, Ali NS, Hussein MA, Thabet GM, azzam OM, Mohamed AY, farghly MI, Al Hussain E, Alkhalil SS, Abouaggour AAM, Ibrahem Fathy Hassan NA, Iqbal S, Mohamed AA, Hafez W, Mahmoud MO. Evaluation of miRNA-146a, miRNA-34a, and pro-inflammatory cytokines as a potential early indicators for type 1 diabetes mellitus. Noncoding RNA Res 2024; 9:1249-1256. [PMID: 39036602 PMCID: PMC11259987 DOI: 10.1016/j.ncrna.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 06/03/2024] [Indexed: 07/23/2024] Open
Abstract
Background Type I diabetes mellitus (T1DM) is one of the most common chronic autoimmune diseases worldwide. miRNAs are a class of small non-coding RNA molecules that have been linked to immune system functions, β-cell metabolism, proliferation, and death, all of which contribute to pathogenesis of TIDM. Dysregulated miRNAs have been identified in Egyptian TIDM patients. Aim Several miRNAs were profiled in Egyptian TIDM patients to determine whether they can be used as molecular biomarkers for T1DM. The relationship between the investigated miRNAs and pro-inflammatory cytokines (TNF-α and IL-6) has also been evaluated in the development of TIDM, in addition to the creation of a proposed model for TIDM prediction. Patients & methods Case-control study included 177 Egyptian patients with confirmed type I diabetes mellitus and 177 healthy individuals. MiRNA-34 and miRNA-146 were detected in serum samples using real-time PCR, whereas TNF-α and IL-6 levels were assessed using ELIZA. Results Patients with TIDM showed a significant decrease in the expression of miRNA-146, with a cut-off value ≤ 3.3, 48 % specificity, and 92.1 % sensitivity, whereas miRNA-34 had the highest sensitivity (95.5 %) and specificity (97.2 %) for differentiating diabetic patients from controls. Furthermore, other diagnostic proinflammatory markers showed lower sensitivity and specificity. Conclusion Serum levels of miRNA-34a, miRNA-146, IL-6, and TNF-α provide new insights into T1DM pathogenesis and could be used for screening and diagnosis purposes. They can be also a potential therapeutic target, as well as allowing for more strategies to improve T1DM disease outcomes.
Collapse
Affiliation(s)
- Amal A. Mohamed
- Biochemistry and Molecular Biology Department, National Hepatology and Tropical Medicine Research Institute, GOTHI, Cairo, Egypt
| | - Gamil M. Abdallah
- Biochemistry Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Ibrahim T. Ibrahim
- Biochemistry Department, Faculty of Pharmacy, Beni Suef University, Beni Suef, Egypt
| | - Nada S. Ali
- Biochemistry Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Mona A. Hussein
- Internal Medicine Department, National Institute of Diabetes and Endocrinology, GOTHI, Cairo, Egypt
| | - Ghada Maher Thabet
- Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Omar M. azzam
- Internal Medicine Department, Ahmed Maher Teaching Hospital, GOTHI, Cairo, Egypt
| | - Amira Yones Mohamed
- Internal medicine department, ELmatareya Teaching Hospital, GOTHI, Cairo, Egypt
| | - Maysa I. farghly
- Department of Clinical Pathology, Faculty of Medicine, Suez University, Suez, Egypt
| | - Eman Al Hussain
- Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Samia S. Alkhalil
- Medical Laboratories Department, College of Applied Medical Sciences in Al Quway'iyah, Shaqraa University, Saudi Arabia
| | | | | | | | | | - Wael Hafez
- Internal Medicine Department, Medical Research and Clinical Studies Institute, The National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, Cairo Governorate 12622, Egypt
| | - Mohamed O. Mahmoud
- Biochemistry Department, Faculty of Pharmacy, Beni Suef University, Beni Suef, Egypt
| |
Collapse
|
6
|
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
|
7
|
Dong H, Sun Y, Nie L, Cui A, Zhao P, Leung WK, Wang Q. Metabolic memory: mechanisms and diseases. Signal Transduct Target Ther 2024; 9:38. [PMID: 38413567 PMCID: PMC10899265 DOI: 10.1038/s41392-024-01755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/29/2024] Open
Abstract
Metabolic diseases and their complications impose health and economic burdens worldwide. Evidence from past experimental studies and clinical trials suggests our body may have the ability to remember the past metabolic environment, such as hyperglycemia or hyperlipidemia, thus leading to chronic inflammatory disorders and other diseases even after the elimination of these metabolic environments. The long-term effects of that aberrant metabolism on the body have been summarized as metabolic memory and are found to assume a crucial role in states of health and disease. Multiple molecular mechanisms collectively participate in metabolic memory management, resulting in different cellular alterations as well as tissue and organ dysfunctions, culminating in disease progression and even affecting offspring. The elucidation and expansion of the concept of metabolic memory provides more comprehensive insight into pathogenic mechanisms underlying metabolic diseases and complications and promises to be a new target in disease detection and management. Here, we retrace the history of relevant research on metabolic memory and summarize its salient characteristics. We provide a detailed discussion of the mechanisms by which metabolic memory may be involved in disease development at molecular, cellular, and organ levels, with emphasis on the impact of epigenetic modulations. Finally, we present some of the pivotal findings arguing in favor of targeting metabolic memory to develop therapeutic strategies for metabolic diseases and provide the latest reflections on the consequences of metabolic memory as well as their implications for human health and diseases.
Collapse
Affiliation(s)
- Hao Dong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuezhang Sun
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lulingxiao Nie
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Aimin Cui
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Pengfei Zhao
- Periodontology and Implant Dentistry Division, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Wai Keung Leung
- Periodontology and Implant Dentistry Division, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Qi Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
8
|
Miron RJ, Estrin NE, Sculean A, Zhang Y. Understanding exosomes: Part 2-Emerging leaders in regenerative medicine. Periodontol 2000 2024; 94:257-414. [PMID: 38591622 DOI: 10.1111/prd.12561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with the ability to communicate with other tissues and cell types over long distances. Their use in regenerative medicine has gained tremendous momentum recently due to their ability to be utilized as therapeutic options for a wide array of diseases/conditions. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be developed. Today exosomes have been applied in numerous contexts including neurodegenerative disorders (Alzheimer's disease, central nervous system, depression, multiple sclerosis, Parkinson's disease, post-traumatic stress disorders, traumatic brain injury, peripheral nerve injury), damaged organs (heart, kidney, liver, stroke, myocardial infarctions, myocardial infarctions, ovaries), degenerative processes (atherosclerosis, diabetes, hematology disorders, musculoskeletal degeneration, osteoradionecrosis, respiratory disease), infectious diseases (COVID-19, hepatitis), regenerative procedures (antiaging, bone regeneration, cartilage/joint regeneration, osteoarthritis, cutaneous wounds, dental regeneration, dermatology/skin regeneration, erectile dysfunction, hair regrowth, intervertebral disc repair, spinal cord injury, vascular regeneration), and cancer therapy (breast, colorectal, gastric cancer and osteosarcomas), immune function (allergy, autoimmune disorders, immune regulation, inflammatory diseases, lupus, rheumatoid arthritis). This scoping review is a first of its kind aimed at summarizing the extensive regenerative potential of exosomes over a broad range of diseases and disorders.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Nathan E Estrin
- Advanced PRF Education, Venice, Florida, USA
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Anton Sculean
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
9
|
Mihanfar A, Akbarzadeh M, Ghazizadeh Darband S, Sadighparvar S, Majidinia M. SIRT1: a promising therapeutic target in type 2 diabetes mellitus. Arch Physiol Biochem 2024; 130:13-28. [PMID: 34379994 DOI: 10.1080/13813455.2021.1956976] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/13/2021] [Indexed: 01/07/2023]
Abstract
A significant increase in the worldwide incidence and prevalence of type 2 diabetic mellitus (T2DM) has elevated the need for studies on novel and effective therapeutic strategies. Sirtuin 1 (SIRT1) is an NAD + dependent protein deacetylase with a critical function in the regulation of glucose/lipid metabolism, insulin resistance, inflammation, oxidative stress, and mitochondrial function. SIRT1 is also involved in the regulation of insulin secretion from pancreatic β-cells and protecting these cells from inflammation and oxidative stress-mediated tissue damages. In this regard, major SIRT1 activators have been demonstrated to exert a beneficial impact in reversing T2DM-related complications including cardiomyopathy, nephropathy, retinopathy, and neuropathy, hence treating T2DM. Therefore, an accumulating number of recent studies have investigated the efficacy of targeting SIRT1 as a therapeutic strategy in T2DM. In this review we aimed to discuss the current understanding of the physiological and biological roles of SIRT1, then its implication in the pathogenesis of T2DM, and the therapeutic potential of SIRT1 in combating T2DM.
Collapse
Affiliation(s)
- Ainaz Mihanfar
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Akbarzadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Shirin Sadighparvar
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
10
|
Chamgordani MK, Bardestani A, Ebrahimpour S, Esmaeili A. In diabetic male Wistar rats, quercetin-conjugated superparamagnetic iron oxide nanoparticles have an effect on the SIRT1/p66Shc-mediated pathway related to cognitive impairment. BMC Pharmacol Toxicol 2023; 24:81. [PMID: 38129872 PMCID: PMC10734159 DOI: 10.1186/s40360-023-00725-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Quercetin (QC) possesses a variety of health-promoting effects in pure and in conjugation with nanoparticles. Since the mRNA-SIRT1/p66Shc pathway and microRNAs (miRNAs) are implicated in the oxidative process, we aimed to compare the effects of QC and QC-conjugated superparamagnetic iron oxide nanoparticles (QCSPIONs) on this pathway. METHODS Through the use of the chemical coprecipitation technique (CPT), SPIONs were synthesized, coated with dextran, and conjugated with quercetin. Adult male Wistar rats were given intraperitoneal injections of streptozotocin to look for signs of type 1 diabetes (T1D). The animals were randomized into five groups: the control group got deionized water (DI), free QC solution (25 mg/kg), SPIONs (25 mg/kg), and QCSPIONs (25 mg/kg), and all groups received repeat doses administered orally over 35 days. Real-time quantitative PCR was used to assess the levels of miR-34a, let-7a-p5, SIRT1, p66Shc, CASP3, and PARP1 expression in the hippocampus of diabetic rats. RESULTS In silico investigations identified p66Shc, CASP3, and PARP1 as targets of let-7a-5p and miR-34a as possible regulators of SIRT1 genes. The outcomes demonstrated that diabetes elevated miR-34a, p66Shc, CASP3, and PARP1 and downregulated let-7a-5p and SIRT1 expression. In contrast to the diabetic group, QCSPIONs boosted let-7a-5p expression levels and consequently lowered p66Shc, CASP3, and PARP1 expression levels. QCSPIONs also reduced miR-34a expression, which led to an upsurge in SIRT1 expression. CONCLUSION Our results suggest that QCSPIONs can regulate the SIRT1/p66Shc-mediated signaling pathway and can be considered a promising candidate for ameliorating the complications of diabetes.
Collapse
Affiliation(s)
- Mahnaz Karami Chamgordani
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, P.O. Box: 8174673441, Iran
| | - Akram Bardestani
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, P.O. Box: 8174673441, Iran
| | - Shiva Ebrahimpour
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, P.O. Box: 8174673441, Iran
| | - Abolghasem Esmaeili
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, P.O. Box: 8174673441, Iran.
| |
Collapse
|
11
|
Biondi G, Marrano N, Borrelli A, Rella M, D’Oria R, Genchi VA, Caccioppoli C, Cignarelli A, Perrini S, Laviola L, Giorgino F, Natalicchio A. The p66 Shc Redox Protein and the Emerging Complications of Diabetes. Int J Mol Sci 2023; 25:108. [PMID: 38203279 PMCID: PMC10778847 DOI: 10.3390/ijms25010108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Diabetes mellitus is a chronic metabolic disease, the prevalence of which is constantly increasing worldwide. It is often burdened by disabling comorbidities that reduce the quality and expectancy of life of the affected individuals. The traditional complications of diabetes are generally described as macrovascular complications (e.g., coronary heart disease, peripheral arterial disease, and stroke), and microvascular complications (e.g., diabetic kidney disease, retinopathy, and neuropathy). Recently, due to advances in diabetes management and the increased life expectancy of diabetic patients, a strong correlation between diabetes and other pathological conditions (such as liver diseases, cancer, neurodegenerative diseases, cognitive impairments, and sleep disorders) has emerged. Therefore, these comorbidities have been proposed as emerging complications of diabetes. P66Shc is a redox protein that plays a role in oxidative stress, apoptosis, glucose metabolism, and cellular aging. It can be regulated by various stressful stimuli typical of the diabetic milieu and is involved in various types of organ and tissue damage under diabetic conditions. Although its role in the pathogenesis of diabetes remains controversial, there is strong evidence regarding the involvement of p66Shc in the traditional complications of diabetes. In this review, we will summarize the evidence supporting the role of p66Shc in the pathogenesis of diabetes and its complications, focusing for the first time on the emerging complications of diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Francesco Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy (M.R.); (R.D.); (V.A.G.)
| | | |
Collapse
|
12
|
Maiese K. Cornerstone Cellular Pathways for Metabolic Disorders and Diabetes Mellitus: Non-Coding RNAs, Wnt Signaling, and AMPK. Cells 2023; 12:2595. [PMID: 37998330 PMCID: PMC10670256 DOI: 10.3390/cells12222595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Metabolic disorders and diabetes (DM) impact more than five hundred million individuals throughout the world and are insidious in onset, chronic in nature, and yield significant disability and death. Current therapies that address nutritional status, weight management, and pharmacological options may delay disability but cannot alter disease course or functional organ loss, such as dementia and degeneration of systemic bodily functions. Underlying these challenges are the onset of aging disorders associated with increased lifespan, telomere dysfunction, and oxidative stress generation that lead to multi-system dysfunction. These significant hurdles point to the urgent need to address underlying disease mechanisms with innovative applications. New treatment strategies involve non-coding RNA pathways with microRNAs (miRNAs) and circular ribonucleic acids (circRNAs), Wnt signaling, and Wnt1 inducible signaling pathway protein 1 (WISP1) that are dependent upon programmed cell death pathways, cellular metabolic pathways with AMP-activated protein kinase (AMPK) and nicotinamide, and growth factor applications. Non-coding RNAs, Wnt signaling, and AMPK are cornerstone mechanisms for overseeing complex metabolic pathways that offer innovative treatment avenues for metabolic disease and DM but will necessitate continued appreciation of the ability of each of these cellular mechanisms to independently and in unison influence clinical outcome.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
13
|
Li Q, Zhang Q. MiR-34a and endothelial biology. Life Sci 2023; 330:121976. [PMID: 37495076 DOI: 10.1016/j.lfs.2023.121976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 07/06/2023] [Accepted: 07/23/2023] [Indexed: 07/28/2023]
Abstract
MicroRNAs (miRNAs) are endogenous ∼22 nt long RNAs that play important gene-regulatory roles in cells by pairing to the mRNAs of protein-coding genes to direct their posttranscriptional repression. Many miRNAs have been identified in endothelial cells and play important roles in endothelial biology. miR-34a is relatively early identified in endothelial cells and has been involved in regulating endothelial functions, angiogenesis, differentiation, senescence, inflammatory response, responses to shear stress, and mitochondrial function. This review outlines the current understanding of miR-34a in endothelial biology and discusses its potential as a therapeutic target to treat vascular diseases.
Collapse
Affiliation(s)
- Qiuxia Li
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA 90095, USA; Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | - Quanjiang Zhang
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
14
|
Li Q, Zhang Q, Kim YR, Gaddam RR, Jacobs JS, Bachschmid MM, Younis T, Zhu Z, Zingman L, London B, Rauckhorst AJ, Taylor EB, Norris AW, Vikram A, Irani K. Deficiency of endothelial sirtuin1 in mice stimulates skeletal muscle insulin sensitivity by modifying the secretome. Nat Commun 2023; 14:5595. [PMID: 37696839 PMCID: PMC10495425 DOI: 10.1038/s41467-023-41351-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 08/31/2023] [Indexed: 09/13/2023] Open
Abstract
Downregulation of endothelial Sirtuin1 (Sirt1) in insulin resistant states contributes to vascular dysfunction. Furthermore, Sirt1 deficiency in skeletal myocytes promotes insulin resistance. Here, we show that deletion of endothelial Sirt1, while impairing endothelial function, paradoxically improves skeletal muscle insulin sensitivity. Compared to wild-type mice, male mice lacking endothelial Sirt1 (E-Sirt1-KO) preferentially utilize glucose over fat, and have higher insulin sensitivity, glucose uptake, and Akt signaling in fast-twitch skeletal muscle. Enhanced insulin sensitivity of E-Sirt1-KO mice is transferrable to wild-type mice via the systemic circulation. Endothelial Sirt1 deficiency, by inhibiting autophagy and activating nuclear factor-kappa B signaling, augments expression and secretion of thymosin beta-4 (Tβ4) that promotes insulin signaling in skeletal myotubes. Thus, unlike in skeletal myocytes, Sirt1 deficiency in the endothelium promotes glucose homeostasis by stimulating skeletal muscle insulin sensitivity through a blood-borne mechanism, and augmented secretion of Tβ4 by Sirt1-deficient endothelial cells boosts insulin signaling in skeletal muscle cells.
Collapse
Affiliation(s)
- Qiuxia Li
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA.
| | - Quanjiang Zhang
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Young-Rae Kim
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Ravinder Reddy Gaddam
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Julia S Jacobs
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | | | - Tsneem Younis
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Zhiyong Zhu
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Veterans Affairs Medical Center, Iowa City, IA, 52242, USA
| | - Leonid Zingman
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Veterans Affairs Medical Center, Iowa City, IA, 52242, USA
| | - Barry London
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Adam J Rauckhorst
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- FOEDRC Metabolomics Core Facility, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Eric B Taylor
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- FOEDRC Metabolomics Core Facility, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Andrew W Norris
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- FOEDRC Metabolic Phenotyping Core Facility, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Ajit Vikram
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Kaikobad Irani
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Veterans Affairs Medical Center, Iowa City, IA, 52242, USA.
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
15
|
Szydełko J, Matyjaszek-Matuszek B. MicroRNAs as Biomarkers for Coronary Artery Disease Related to Type 2 Diabetes Mellitus-From Pathogenesis to Potential Clinical Application. Int J Mol Sci 2022; 24:ijms24010616. [PMID: 36614057 PMCID: PMC9820734 DOI: 10.3390/ijms24010616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease with still growing incidence among adults and young people worldwide. Patients with T2DM are more susceptible to developing coronary artery disease (CAD) than non-diabetic individuals. The currently used diagnostic methods do not ensure the detection of CAD at an early stage. Thus, extensive research on non-invasive, blood-based biomarkers is necessary to avoid life-threatening events. MicroRNAs (miRNAs) are small, endogenous, non-coding RNAs that are stable in human body fluids and easily detectable. A number of reports have highlighted that the aberrant expression of miRNAs may impair the diversity of signaling pathways underlying the pathophysiology of atherosclerosis, which is a key player linking T2DM with CAD. The preclinical evidence suggests the atheroprotective and atherogenic influence of miRNAs on every step of T2DM-induced atherogenesis, including endothelial dysfunction, endothelial to mesenchymal transition, macrophage activation, vascular smooth muscle cells proliferation/migration, platelet hyperactivity, and calcification. Among the 122 analyzed miRNAs, 14 top miRNAs appear to be the most consistently dysregulated in T2DM and CAD, whereas 10 miRNAs are altered in T2DM, CAD, and T2DM-CAD patients. This up-to-date overview aims to discuss the role of miRNAs in the development of diabetic CAD, emphasizing their potential clinical usefulness as novel, non-invasive biomarkers and therapeutic targets for T2DM individuals with a predisposition to undergo CAD.
Collapse
|
16
|
Mousavi S, Khazeei Tabari MA, Bagheri A, Samieefar N, Shaterian N, Kelishadi R. The Role of p66Shc in Diabetes: A Comprehensive Review from Bench to Bedside. J Diabetes Res 2022; 2022:7703520. [PMID: 36465704 PMCID: PMC9715346 DOI: 10.1155/2022/7703520] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022] Open
Abstract
It is well-documented that diabetes is an inflammatory and oxidative disease, with an escalating global burden. Still, there is no definite treatment for diabetes or even prevention of its harmful complications. Therefore, understanding the molecular pathways associated with diabetes might help in finding a solution. p66Shc is a member of Shc family proteins, and it is considered as an oxidative stress sensor and regulator in cells. There are inconsistent data about the role of p66Shc in inducing diabetes, but accumulating evidence supports its role in the pathogenesis of diabetes-related complications, including macro and microangiopathies. There is growing hope that by understanding and targeting molecular pathways involved in this network, prevention of diabetes or its complications would be achievable. This review provides an overview about the role of p66Shc in the development of diabetes and its complications.
Collapse
Affiliation(s)
- SeyedehFatemeh Mousavi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, Mazandaran University of Medical Sciences, Mazandaran, Iran
- USERN Office, Mazandaran University of Medical Sciences, Mazandaran, Iran
| | - Alireza Bagheri
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Noosha Samieefar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Negar Shaterian
- Student Research Committee, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- USERN Office, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Roya Kelishadi
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
- USERN Office, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
17
|
Nappi F, Fiore A, Masiglat J, Cavuoti T, Romandini M, Nappi P, Avtaar Singh SS, Couetil JP. Endothelium-Derived Relaxing Factors and Endothelial Function: A Systematic Review. Biomedicines 2022; 10:2884. [PMID: 36359402 PMCID: PMC9687749 DOI: 10.3390/biomedicines10112884] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/05/2022] [Accepted: 11/06/2022] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND The endothelium plays a pivotal role in homeostatic mechanisms. It specifically modulates vascular tone by releasing vasodilatory mediators, which act on the vascular smooth muscle. Large amounts of work have been dedicated towards identifying mediators of vasodilation and vasoconstriction alongside the deleterious effects of reactive oxygen species on the endothelium. We conducted a systematic review to study the role of the factors released by the endothelium and the effects on the vessels alongside its role in atherosclerosis. METHODS A search was conducted with appropriate search terms. Specific attention was offered to the effects of emerging modulators of endothelial functions focusing the analysis on studies that investigated the role of reactive oxygen species (ROS), perivascular adipose tissue, shear stress, AMP-activated protein kinase, potassium channels, bone morphogenic protein 4, and P2Y2 receptor. RESULTS 530 citations were reviewed, with 35 studies included in the final systematic review. The endpoints were evaluated in these studies which offered an extensive discussion on emerging modulators of endothelial functions. Specific factors such as reactive oxygen species had deleterious effects, especially in the obese and elderly. Another important finding included the shear stress-induced endothelial nitric oxide (NO), which may delay development of atherosclerosis. Perivascular Adipose Tissue (PVAT) also contributes to reparative measures against atherosclerosis, although this may turn pathological in obese subjects. Some of these factors may be targets for pharmaceutical agents in the near future. CONCLUSION The complex role and function of the endothelium is vital for regular homeostasis. Dysregulation may drive atherogenesis; thus, efforts should be placed at considering therapeutic options by targeting some of the factors noted.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| | - Antonio Fiore
- Department of Cardiac Surgery, Hôpitaux Universitaires Henri Mondor, Assistance Publique-Hôpitaux de Paris, 94000 Creteil, France
| | - Joyce Masiglat
- Department of Cardiac Surgery, Hôpitaux Universitaires Henri Mondor, Assistance Publique-Hôpitaux de Paris, 94000 Creteil, France
| | - Teresa Cavuoti
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| | - Michela Romandini
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| | - Pierluigi Nappi
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | | | - Jean-Paul Couetil
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
18
|
Li T, Yu X, Zhu X, Wen Y, Zhu M, Cai W, Hou B, Xu F, Qiu L. Vaccarin alleviates endothelial inflammatory injury in diabetes by mediating miR-570-3p/HDAC1 pathway. Front Pharmacol 2022; 13:956247. [PMID: 36120375 PMCID: PMC9475173 DOI: 10.3389/fphar.2022.956247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Vaccarin is a flavonoid glycoside, which has a variety of pharmacological properties and plays a protective role in diabetes and its complications, but its mechanism is unclear. In this study, we aim to investigate whether histone deacetylase 1(HDAC1), a gene that plays a pivotal role in regulating eukaryotic gene expression, is the target of miR-570-3p in diabetic vascular endothelium, and the potential molecular mechanism of vaccarin regulating endothelial inflammatory injury through miR-570-3p/HDAC1 pathway. The HFD and streptozotocin (STZ) induced diabetes mice model, a classical type 2 diabetic model, was established. The aorta of diabetic mice displayed a decrease of miR-570-3p, the elevation of HDAC1, and inflammatory injury, which were alleviated by vaccarin. Next, we employed the role of vaccarin in regulating endothelial cells miR-570-3p and HDAC1 under hyperglycemia conditions in vitro. We discovered that overexpression of HDAC1 counteracted the inhibitory effect of vaccarin on inflammatory injury in human umbilical vein endothelial cells (HUVECs). Manipulation of miRNA levels in HUVECs was achieved by transfecting cells with miR-570-3p mimic and inhibitor. Overexpression of miR-570-3p could decrease the expression of downstream components of HDAC1 including TNF-α, IL-1β, and malondialdehyde, while increasing GSH-Px activity in HUVECs under hyperglycemic conditions. Nevertheless, such phenomenon was completely reversed by miR-570-3p inhibitor, and administration of miR-570-3p inhibitor could block the inhibition of vaccarin on HDAC1 and inflammatory injury. Luciferase reporter assay confirmed the 3′- UTR of the HDAC1 gene was a direct target of miR-570-3p. In summary, our findings suggest that vaccarin alleviates endothelial inflammatory injury in diabetes by mediating miR-570-3p/HDAC1 pathway. Our study provides a new pathogenic link between deregulation of miRNA expression in the vascular endothelium of diabetes and inflammatory injury and provides new ideas, insights, and choices for the scope of application and medicinal value of vaccarin and some potential biomarkers or targets in diabetic endothelial dysfunction and vascular complications.
Collapse
Affiliation(s)
- Taiyue Li
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaoyi Yu
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu, China
| | - Xuerui Zhu
- School of Life Science and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Yuanyuan Wen
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu, China
| | - Meizhen Zhu
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu, China
| | - Weiwei Cai
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu, China
| | - Bao Hou
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu, China
| | - Fei Xu
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu, China
- *Correspondence: Fei Xu, ; Liying Qiu,
| | - Liying Qiu
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu, China
- *Correspondence: Fei Xu, ; Liying Qiu,
| |
Collapse
|
19
|
Chen X, Shi C, Wang Y, Yu H, Zhang Y, Zhang J, Li P, Gao J. The mechanisms of glycolipid metabolism disorder on vascular injury in type 2 diabetes. Front Physiol 2022; 13:952445. [PMID: 36117707 PMCID: PMC9473659 DOI: 10.3389/fphys.2022.952445] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with diabetes have severe vascular complications, such as diabetic nephropathy, diabetic retinopathy, cardiovascular disease, and neuropathy. Devastating vascular complications lead to increased mortality, blindness, kidney failure, and decreased overall quality of life in people with type 2 diabetes (T2D). Glycolipid metabolism disorder plays a vital role in the vascular complications of T2D. However, the specific mechanism of action remains to be elucidated. In T2D patients, vascular damage begins to develop before insulin resistance and clinical diagnosis. Endothelial dysregulation is a significant cause of vascular complications and the early event of vascular injury. Hyperglycemia and hyperlipidemia can trigger inflammation and oxidative stress, which impair endothelial function. Furthermore, during the pathogenesis of T2D, epigenetic modifications are aberrant and activate various biological processes, resulting in endothelial dysregulation. In the present review, we provide an overview and discussion of the roles of hyperglycemia- and hyperlipidemia-induced endothelial dysfunction, inflammatory response, oxidative stress, and epigenetic modification in the pathogenesis of T2D. Understanding the connections of glucotoxicity and lipotoxicity with vascular injury may reveal a novel potential therapeutic target for diabetic vascular complications.
Collapse
Affiliation(s)
- Xiatian Chen
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | | | - Yin Wang
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Hua Yu
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| | - Yu Zhang
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jiaxuan Zhang
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Peifeng Li
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jinning Gao
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
20
|
Cheng CK, Shang W, Liu J, Cheang WS, Wang Y, Xiang L, Lau CW, Luo JY, Ng CF, Huang Y, Wang L. Activation of AMPK/miR-181b Axis Alleviates Endothelial Dysfunction and Vascular Inflammation in Diabetic Mice. Antioxidants (Basel) 2022; 11:1137. [PMID: 35740034 PMCID: PMC9220246 DOI: 10.3390/antiox11061137] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 02/01/2023] Open
Abstract
Hyperglycemia in diabetes mellitus impairs endothelial function and disrupts microRNA (miRNA) profiles in vasculature, increasing the risk of diabetes-associated complications, including coronary artery disease, diabetic retinopathy, and diabetic nephropathy. miR-181b was previously reported to be an anti-inflammatory mediator in vasculature against atherosclerosis. The current study aimed to investigate whether miR-181b ameliorates diabetes-associated endothelial dysfunction, and to identify potential molecular mechanisms and upstream inducer of miR-181b. We found that miR-181b level was decreased in renal arteries of diabetic patients and in advanced glycation end products (AGEs)-treated renal arteries of non-diabetic patients. Transfection of miR-181b mimics improved endothelium-dependent vasodilation in aortas of high fat diet (HFD)/streptozotocin (STZ)-induced diabetic mice, accompanied by suppression of superoxide overproduction and vascular inflammation markers. AMPK activator-induced AMPK activation upregulated miR-181b level in human umbilical vein endothelial cells (HUVECs). Chronic exercise, potentially through increased blood flow, activated AMPK/miR-181b axis in aortas of diabetic mice. Exposure to laminar shear stress upregulated miR-181b expression in HUVECs. Overall, our findings highlight a critical role of AMPK/miR-181b axis and extend the benefits of chronic exercise in counteracting diabetes-associated endothelial dysfunction.
Collapse
Affiliation(s)
- Chak-Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong 999077, China;
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.); (Y.W.); (L.X.); (C.-W.L.); (J.-Y.L.)
| | - Wenbin Shang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.); (Y.W.); (L.X.); (C.-W.L.); (J.-Y.L.)
| | - Jian Liu
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100871, China;
| | - Wai-San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Zhuhai 519000, China;
| | - Yu Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.); (Y.W.); (L.X.); (C.-W.L.); (J.-Y.L.)
| | - Li Xiang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.); (Y.W.); (L.X.); (C.-W.L.); (J.-Y.L.)
| | - Chi-Wai Lau
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.); (Y.W.); (L.X.); (C.-W.L.); (J.-Y.L.)
| | - Jiang-Yun Luo
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.); (Y.W.); (L.X.); (C.-W.L.); (J.-Y.L.)
| | - Chi-Fai Ng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong 999077, China;
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong 999077, China;
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.); (Y.W.); (L.X.); (C.-W.L.); (J.-Y.L.)
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong 999077, China;
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.); (Y.W.); (L.X.); (C.-W.L.); (J.-Y.L.)
| |
Collapse
|
21
|
Dhawan P, Vasishta S, Balakrishnan A, Joshi MB. Mechanistic insights into glucose induced vascular epigenetic reprogramming in type 2 diabetes. Life Sci 2022; 298:120490. [DOI: 10.1016/j.lfs.2022.120490] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/22/2022] [Accepted: 03/16/2022] [Indexed: 12/13/2022]
|
22
|
Zhang L, Guo Y, Shi S, Zhuge Y, Chen N, Ding Z, Jin B. Tetrahydroxy stilbene glycoside attenuates endothelial cell premature senescence induced by H 2O 2 through the microRNA-34a/SIRT1 pathway. Sci Rep 2022; 12:1708. [PMID: 35105933 PMCID: PMC8807705 DOI: 10.1038/s41598-022-05804-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 01/18/2022] [Indexed: 11/09/2022] Open
Abstract
Numerous studies have demonstrated that endothelial cell senescence plays a decisive role in the development and progression of cardiovascular diseases (CVD). Our previous results confirmed that Tetrahydroxy stilbene glycoside (TSG) can alleviate the human umbilical vein endothelial cells (HUVECs) senescence induced by H2O2 through SIRT1. It has been reported that miR-34a is a translational suppressor of SIRT1. In this study, we aimed to explore whether TSG regulates SIRT1 through miR-34a to ameliorate HUVECs senescence. H2O2 was used to induce premature senescence in HUVECs, and miR-34a mimic or inhibitor were transfected to over-express or suppress the expression level of miR-34a. Results revealed that TSG apparently decreased the miR-34a expression level in H2O2-induced premature senescence of HUVECs. When SIRT1 expression was inhibited by EX527, the attenuation of TSG on the expression level of miR-34a were abolished. When miR-34a expression was knockdown, the effect of TSG on HUVECs senescence could be enhanced. While miR-34a mimic could reverse the effect of TSG on HUVECs senescence. In conclusion, we demonstrated that TSG could attenuated endothelial cell senescence by targeting miR-34a/SIRT1 pathway.
Collapse
Affiliation(s)
- Lixuan Zhang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Yan Guo
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Shennan Shi
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Yani Zhuge
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Nipi Chen
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Bo Jin
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
23
|
Mone P, de Donato A, Varzideh F, Kansakar U, Jankauskas SS, Pansini A, Santulli G. Functional role of miR-34a in diabetes and frailty. FRONTIERS IN AGING 2022; 3:949924. [PMID: 35923683 PMCID: PMC9340262 DOI: 10.3389/fragi.2022.949924] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 06/29/2022] [Indexed: 01/05/2023]
Abstract
Emerging evidence has shown that microRNAs (miRNAs) play critical role in the pathogenesis of several disorders. In the present minireview, we focus our attention on the functional role of a specific miRNA, namely miR-34a, in the pathophysiology of frailty and diabetes mellitus. Based on the current literature, we speculate that this miRNA may serve as a potential biomarker of frailty in diabetic older adults. Additionally, its actions on oxidative stress might represent a druggable target to obtain new potentials treatments.
Collapse
Affiliation(s)
- Pasquale Mone
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
- ASL Avellino, Avellino, Italy
- *Correspondence: Pasquale Mone, ;,
| | | | - Fahimeh Varzideh
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | - Urna Kansakar
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | - Stanislovas S. Jankauskas
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | | | - Gaetano Santulli
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
- Department of Molecular Pharmacology, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
24
|
Fluitt MB, Mohit N, Gambhir KK, Nunlee-Bland G. To the Future: The Role of Exosome-Derived microRNAs as Markers, Mediators, and Therapies for Endothelial Dysfunction in Type 2 Diabetes Mellitus. J Diabetes Res 2022; 2022:5126968. [PMID: 35237694 PMCID: PMC8885279 DOI: 10.1155/2022/5126968] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/08/2022] [Indexed: 01/19/2023] Open
Abstract
The prevalence of diabetes mellitus (DM) is increasing at a staggering rate around the world. In the United States, more than 30.3 million Americans have DM. Type 2 diabetes mellitus (T2DM) accounts for 91.2% of diabetic cases and disproportionately affects African Americans and Hispanics. T2DM is a major risk factor for cardiovascular disease (CVD) and is the leading cause of morbidity and mortality among diabetic patients. While significant advances in T2DM treatment have been made, intensive glucose control has failed to reduce the development of macro and microvascular related deaths in this group. This highlights the need to further elucidate the underlying molecular mechanisms contributing to CVD in the setting of T2DM. Endothelial dysfunction (ED) plays an important role in the development of diabetes-induced vascular complications, including CVD and diabetic nephropathy (DN). Thus, the endothelium provides a lucrative means to investigate the molecular events involved in the development of vascular complications associated with T2DM. microRNAs (miRNA) participate in numerous cellular responses, including mediating messages in vascular homeostasis. Exosomes are small extracellular vesicles (40-160 nanometers) that are abundant in circulation and can deliver various molecules, including miRNAs, from donor to recipient cells to facilitate cell-to-cell communication. Endothelial cells are in constant contact with exosomes (and exosomal content) that can induce a functional response. This review discusses the modulatory role of exosomal miRNAs and proteins in diabetes-induced endothelial dysfunction, highlighting the significance of miRNAs as markers, mediators, and potential therapeutic interventions to ameliorate ED in this patient group.
Collapse
Affiliation(s)
- Maurice B. Fluitt
- Division of Endocrinology and Metabolism, Department of Medicine, Howard University College of Medicine, 520 W St NW, Washington, DC 20059, USA
| | - Neal Mohit
- Division of Endocrinology and Metabolism, Department of Medicine, Howard University College of Medicine, 520 W St NW, Washington, DC 20059, USA
- Department of Biology, Howard University, 415 College St. NW, Washington, DC 20059, USA
| | - Kanwal K. Gambhir
- Division of Endocrinology and Metabolism, Department of Medicine, Howard University College of Medicine, 520 W St NW, Washington, DC 20059, USA
| | - Gail Nunlee-Bland
- Division of Endocrinology and Metabolism, Department of Medicine, Howard University College of Medicine, 520 W St NW, Washington, DC 20059, USA
- Diabetes Treatment Center, Howard University Hospital, 2041 Georgia Ave, NW, Washington, DC 20060, USA
| |
Collapse
|
25
|
Begum MK, Konja D, Singh S, Chlopicki S, Wang Y. Endothelial SIRT1 as a Target for the Prevention of Arterial Aging: Promises and Challenges. J Cardiovasc Pharmacol 2021; 78:S63-S77. [PMID: 34840264 DOI: 10.1097/fjc.0000000000001154] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 09/25/2021] [Indexed: 12/15/2022]
Abstract
ABSTRACT SIRT1, a member of the sirtuin family of longevity regulators, possesses potent activities preventing vascular aging. The expression and function of SIRT1 in endothelial cells are downregulated with age, in turn causing early vascular aging and predisposing various vascular abnormalities. Overexpression of SIRT1 in the vascular endothelium prevents aging-associated endothelial dysfunction and senescence, thus the development of hypertension and atherosclerosis. Numerous efforts have been directed to increase SIRT1 signaling as a potential strategy for different aging-associated diseases. However, the complex mechanisms underlying the regulation of SIRT1 have posed a significant challenge toward the design of specific and effective therapeutics. This review aimed to provide a summary on the regulation and function of SIRT1 in the vascular endothelium and to discuss the different approaches targeting this molecule for the prevention and treatment of age-related cardiovascular and cerebrovascular diseases.
Collapse
Affiliation(s)
- Musammat Kulsuma Begum
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Daniels Konja
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Sandeep Singh
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland; and
- Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
26
|
Abstract
Sirtuin1 is a nutrient-sensitive class III histone deacetylase which is a well-known regulator of organismal lifespan. It has been extensively studied for its role in metabolic regulation as well. Along with its involvement in ageing and metabolism, Sirtuin1 directly deacetylates many critical proteins controlling cardiovascular pathophysiology. Studies using conditional expression and deletion of Sirtuin1 have revealed that it functions in a highly tissue/organ-specific manner. In the vasculature, Sirtuin1 controls endothelial homoeostasis by governing the expression of inflammatory mediators, oxidants and essential transcription factors. Adding to this complexity, Sirtuin1 expression and/or function is also governed by some of these target proteins. Therefore, the importance of better understanding the organ and tissue specificity of Sirtuin1 is highly desirable. Considering the huge volume of research done in this field, this review focuses on Sirtuin1 targets regulating vascular endothelial function. Here, we summarize the discovery of Sirtuin1 as a transcription controller and the further identification of direct target proteins involved in the vascular physiology. Overall, this review presents a holistic picture of the complex cross-talk involved in the molecular regulation of vascular physiology by Sirtuin1.
Collapse
Affiliation(s)
- Jitendra Kumar
- François M. Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Santosh Kumar
- François M. Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
27
|
Theofilis P, Sagris M, Oikonomou E, Antonopoulos AS, Siasos G, Tsioufis C, Tousoulis D. Inflammatory Mechanisms Contributing to Endothelial Dysfunction. Biomedicines 2021; 9:781. [PMID: 34356845 PMCID: PMC8301477 DOI: 10.3390/biomedicines9070781] [Citation(s) in RCA: 280] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 06/30/2021] [Accepted: 07/04/2021] [Indexed: 12/16/2022] Open
Abstract
Maintenance of endothelial cell integrity is an important component of human health and disease since the endothelium can perform various functions including regulation of vascular tone, control of hemostasis and thrombosis, cellular adhesion, smooth muscle cell proliferation, and vascular inflammation. Endothelial dysfunction is encompassed by complex pathophysiology that is based on endothelial nitric oxide synthase uncoupling and endothelial activation following stimulation from various inflammatory mediators (molecular patterns, oxidized lipoproteins, cytokines). The downstream signaling via nuclear factor-κB leads to overexpression of adhesion molecules, selectins, and chemokines that facilitate leukocyte adhesion, rolling, and transmigration to the subendothelial space. Moreover, oscillatory shear stress leads to pro-inflammatory endothelial activation with increased monocyte adhesion and endothelial cell apoptosis, an effect that is dependent on multiple pathways and flow-sensitive microRNA regulation. Moreover, the role of neutrophil extracellular traps and NLRP3 inflammasome as inflammatory mechanisms contributing to endothelial dysfunction has recently been unveiled and is under further investigation. Consequently, and following their activation, injured endothelial cells release inflammatory mediators and enter a pro-thrombotic state through activation of coagulation pathways, downregulation of thrombomodulin, and an increase in platelet adhesion and aggregation owing to the action of von-Willebrand factor, ultimately promoting atherosclerosis progression.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
| | - Marios Sagris
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
| | - Evangelos Oikonomou
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
- 3rd Department of Cardiology, Thoracic Diseases General Hospital Sotiria, University of Athens Medical School, 11527 Athens, Greece
| | - Alexios S. Antonopoulos
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
| | - Gerasimos Siasos
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
- 3rd Department of Cardiology, Thoracic Diseases General Hospital Sotiria, University of Athens Medical School, 11527 Athens, Greece
| | - Costas Tsioufis
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
| | - Dimitris Tousoulis
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
| |
Collapse
|
28
|
Liang Y, Liu H, Zhu J, Song N, Lu Z, Fang Y, Teng J, Dai Y, Ding X. Inhibition of p53/miR-34a/SIRT1 axis ameliorates podocyte injury in diabetic nephropathy. Biochem Biophys Res Commun 2021; 559:48-55. [PMID: 33932899 DOI: 10.1016/j.bbrc.2021.04.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 10/21/2022]
Abstract
Podocyte injury is associated with albuminuria and the progression of diabetic nephropathy (DN). MiR-34a, a p53-regulated miRNA, directly targets SIRT1 and contributed to DN progression. MiR-34a represses SIRT1 to activate p53 and establish a positive feedback loop. However, whether p53/miR-34a/SIRT1 signaling is activated in podocytes and contributes to DN pathogenesis remains elusive. In this study, we observed that serum miR-34a level was positively correlated with podocyte injury in DN patients. The expression of acetylated p53 and miR-34a was upregulated, SIRT1was downregulated in glomeruli from patients with DN and STZ induced diabetic mice, as well as in human podocytes treated with advanced glycation end (AGE). MiR-34a antagonism in vitro and vivo in STZ induced diabetic mice developed alleviated glomerulus injury as reflected by attenuated albuminuria, reduced podocyte loss and restored autophagic flux. In human podocyte, inhibition of AGE formation by pyridoxamine prevented miR-34a dependent repression of SIRT1, p53 acetylation and activate podocyte autophagy in a dose-dependent manner. MiR-34a overexpression increases acetylation of p53 by translational repression of SIRT1. SIRT1 overexpression also impacts AGE induced apoptosis through deacetylating p53, whereas silencing of SIRT1 by EX527 attenuated the cytoprotective functions of miR-34a knockdown. Moreover, blockade of p53 acetylation significantly rescued miR-34a-induced apoptosis through SIRT1 restoration. Collectively, we demonstrate that by activation of p53, AGE induced the transcription of miR-34a, miR-34a in turn repressed SIRT1 to activate p53, resulting in a positive-feedback loop and contributing to podocyte injury. Targeting modulation of p53/miR-34a/SIRT1 feedback by miR-34a knockdown or overexpression of SIRT1 could rescue podocyte injury during DN.
Collapse
Affiliation(s)
- Yiran Liang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| | - Hong Liu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| | - Jiaming Zhu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| | - Nana Song
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| | - Zhihui Lu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| | - Yi Fang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| | - Jie Teng
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| | - Yan Dai
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China.
| | - Xiaoqiang Ding
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China.
| |
Collapse
|
29
|
Hamdani N, Costantino S, Mügge A, Lebeche D, Tschöpe C, Thum T, Paneni F. Leveraging clinical epigenetics in heart failure with preserved ejection fraction: a call for individualized therapies. Eur Heart J 2021; 42:1940-1958. [PMID: 36282124 DOI: 10.1093/eurheartj/ehab197] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/17/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022] Open
Abstract
Described as the 'single largest unmet need in cardiovascular medicine', heart failure with preserved ejection fraction (HFpEF) remains an untreatable disease currently representing 65% of new heart failure diagnoses. HFpEF is more frequent among women and associates with a poor prognosis and unsustainable healthcare costs. Moreover, the variability in HFpEF phenotypes amplifies complexity and difficulties in the approach. In this perspective, unveiling novel molecular targets is imperative. Epigenetic modifications-defined as changes of DNA, histones, and non-coding RNAs (ncRNAs)-represent a molecular framework through which the environment modulates gene expression. Epigenetic signals acquired over the lifetime lead to chromatin remodelling and affect transcriptional programmes underlying oxidative stress, inflammation, dysmetabolism, and maladaptive left ventricular remodelling, all conditions predisposing to HFpEF. The strong involvement of epigenetic signalling in this setting makes the epigenetic information relevant for diagnostic and therapeutic purposes in patients with HFpEF. The recent advances in high-throughput sequencing, computational epigenetics, and machine learning have enabled the identification of reliable epigenetic biomarkers in cardiovascular patients. Contrary to genetic tools, epigenetic biomarkers mirror the contribution of environmental cues and lifestyle changes and their reversible nature offers a promising opportunity to monitor disease states. The growing understanding of chromatin and ncRNAs biology has led to the development of several Food and Drug Administration approved 'epidrugs' (chromatin modifiers, mimics, anti-miRs) able to prevent transcriptional alterations underpinning left ventricular remodelling and HFpEF. In the present review, we discuss the importance of clinical epigenetics as a new tool to be employed for a personalized management of HFpEF.
Collapse
Affiliation(s)
- Nazha Hamdani
- Institute of Physiology, Ruhr University, Bochum, Germany.,Molecular and Experimental Cardiology, Ruhr University, Bochum, Germany.,Department of Cardiology, St-Josef Hospital, Ruhr University, Bochum, Germany.,Clinical Pharmacology, Ruhr University, Bochum, Germany
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren CH-8952, Switzerland
| | - Andreas Mügge
- Molecular and Experimental Cardiology, Ruhr University, Bochum, Germany.,Department of Cardiology, St-Josef Hospital, Ruhr University, Bochum, Germany
| | - Djamel Lebeche
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY 10029, USA.,Department of Medicine, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Medicine, Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany.,Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany
| | - 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 (ITEM), Hannover 30625, Germany
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren CH-8952, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Zürich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zürich, Switzerland
| |
Collapse
|
30
|
MicroRNAs and Oxidative Stress: An Intriguing Crosstalk to Be Exploited in the Management of Type 2 Diabetes. Antioxidants (Basel) 2021; 10:antiox10050802. [PMID: 34069422 PMCID: PMC8159096 DOI: 10.3390/antiox10050802] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 11/16/2022] Open
Abstract
Type 2 diabetes is a chronic disease widespread throughout the world, with significant human, social, and economic costs. Its multifactorial etiology leads to persistent hyperglycemia, impaired carbohydrate and fat metabolism, chronic inflammation, and defects in insulin secretion or insulin action, or both. Emerging evidence reveals that oxidative stress has a critical role in the development of type 2 diabetes. Overproduction of reactive oxygen species can promote an imbalance between the production and neutralization of antioxidant defence systems, thus favoring lipid accumulation, cellular stress, and the activation of cytosolic signaling pathways, and inducing β-cell dysfunction, insulin resistance, and tissue inflammation. Over the last few years, microRNAs (miRNAs) have attracted growing attention as important mediators of diverse aspects of oxidative stress. These small endogenous non-coding RNAs of 19-24 nucleotides act as negative regulators of gene expression, including the modulation of redox signaling pathways. The present review aims to provide an overview of the current knowledge concerning the molecular crosstalk that takes place between oxidative stress and microRNAs in the physiopathology of type 2 diabetes, with a special emphasis on its potential as a therapeutic target.
Collapse
|
31
|
Feng J, Bao L, Wang X, Li H, Chen Y, Xiao W, Li Z, Xie L, Lu W, Jiang H, Lee K, He JC. Low expression of HIV genes in podocytes accelerates the progression of diabetic kidney disease in mice. Kidney Int 2021; 99:914-925. [PMID: 33359498 PMCID: PMC8006538 DOI: 10.1016/j.kint.2020.12.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 11/11/2020] [Accepted: 12/07/2020] [Indexed: 01/22/2023]
Abstract
With the widespread use combination antiretroviral therapy, there has been a dramatic decrease in HIV-associated nephropathy. However, although the patients living with HIV have low or undetectable viral load, the prevalence of chronic kidney disease (CKD) in this population remains high. Additionally, improved survival is associated with aging-related comorbidities such as diabetes and cardiovascular disease. A faster progression of CKD is associated with concurrent HIV infection and diabetes than with HIV infection or diabetes alone. To explore the potential pathogenic mechanisms that synergistically drive CKD progression by diabetes and HIV infection, we generated a new mouse model with a relatively low expression of HIV-1 proviral genes specifically in podocytes (pod-HIV mice) to better mimic the setting of kidney injury in patients living with HIV. While no apparent kidney phenotypes were observed at baseline in pod-HIV mice, the induction of mild diabetic kidney disease with streptozotocin led to significant worsening of albuminuria, glomerular injury, podocyte loss, and kidney dysfunction as compared to the mice with diabetes alone. Mechanistically, diabetes and HIV-1 synergistically increased the glomerular expression of microRNA-34a (miR-34a), thereby reducing the expression of Sirtuin-1 (SIRT1) deacetylase. These changes were also associated with increased acetylation and activation of p53 and p65 NF-κB and with enhanced expression of senescence and inflammatory markers. The treatment of diabetic pod-HIV mice with the specific Sirtuin-1 agonist BF175 significantly attenuated albuminuria and glomerulopathy. Thus, our study highlights the reduction in Sirtuin-1 as a major basis of CKD progression in diabetic patients living with HIV and suggests Sirtuin-1 agonists as a potential therapy.
Collapse
Affiliation(s)
- Jie Feng
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Bao
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Xuan Wang
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Huilin Li
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Yuqiang Chen
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Wenzhen Xiao
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Zhengzhe Li
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Liyi Xie
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wanhong Lu
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hongli Jiang
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kyung Lee
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, USA.
| | - John Cijiang He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, USA; Renal Section, James J. Peters VAMC, Bronx, New York, USA.
| |
Collapse
|
32
|
Lee Y, Im E. Regulation of miRNAs by Natural Antioxidants in Cardiovascular Diseases: Focus on SIRT1 and eNOS. Antioxidants (Basel) 2021; 10:antiox10030377. [PMID: 33802566 PMCID: PMC8000568 DOI: 10.3390/antiox10030377] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of morbidity and mortality worldwide. The potential benefits of natural antioxidants derived from supplemental nutrients against CVDs are well known. Remarkably, natural antioxidants exert cardioprotective effects by reducing oxidative stress, increasing vasodilation, and normalizing endothelial dysfunction. Recently, considerable evidence has highlighted an important role played by the synergistic interaction between endothelial nitric oxide synthase (eNOS) and sirtuin 1 (SIRT1) in the maintenance of endothelial function. To provide a new perspective on the role of natural antioxidants against CVDs, we focused on microRNAs (miRNAs), which are important posttranscriptional modulators in human diseases. Several miRNAs are regulated via the consumption of natural antioxidants and are related to the regulation of oxidative stress by targeting eNOS and/or SIRT1. In this review, we have discussed the specific molecular regulation of eNOS/SIRT1-related endothelial dysfunction and its contribution to CVD pathologies; furthermore, we selected nine different miRNAs that target the expression of eNOS and SIRT1 in CVDs. Additionally, we have summarized the alteration of miRNA expression and regulation of activities of miRNA through natural antioxidant consumption.
Collapse
Affiliation(s)
| | - Eunok Im
- Correspondence: ; Tel.: +82-51-510-2812; Fax: +82-51-513-6754
| |
Collapse
|
33
|
Xu Y, Zhang Y, Liang H, Liu X. Coumestrol mitigates retinal cell inflammation, apoptosis, and oxidative stress in a rat model of diabetic retinopathy via activation of SIRT1. Aging (Albany NY) 2021; 13:5342-5357. [PMID: 33536350 PMCID: PMC7950241 DOI: 10.18632/aging.202467] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022]
Abstract
Diabetes-induced oxidative stress is vital in initiating neuronal damage in the diabetic retina, leading to diabetic retinopathy (DR). This study investigates the possible effects of coumestrol (CMS) on streptozotocin (STZ)-induced DR. First, we established a rat model of DR by STZ injection and a cell model involving high-glucose (HG) exposure of human retinal microvascular endothelial cells (hRMECs). We characterized the expression patterns of oxidative stress indicators, pro-inflammatory cytokines, and pro-apoptotic proteins in hRMECs. Polymerase chain reaction showed sirtuin 1 (SIRT1) to be poorly expressed in the retinal tissues of STZ-treated rats and HG-exposed hRMECs, but its expression was upregulated upon treatment with CMS treatment. Furthermore, CMS treatment attenuated the STZ-induced pathologies such as oxidative stress, inflammation, and cell apoptosis. Consistent with the in vivo results, CMS activated the expression of SIRT1, thereby inhibiting oxidative stress, inflammation, and apoptosis of HG-treated hRMECs. From these findings, we concluded that CMS ameliorated DR by inhibiting inflammation, apoptosis and oxidative stress through activation of SIRT1.
Collapse
Affiliation(s)
- Yanchao Xu
- The Second Ward, Department of Endocrinology and Metabolism, Linyi People's Hospital of Shandong Province, Linyi 276000, P. R. China
| | - Yusong Zhang
- Imaging Center, Linyi People's Hospital of Shandong Province, Linyi 276000, P. R. China
| | - Hongwei Liang
- Department of Health Care, Linyi People's Hospital of Shandong Province, Linyi 276000, P. R. China
| | - Xiaomeng Liu
- The Second Ward, Department of Endocrinology and Metabolism, Linyi People's Hospital of Shandong Province, Linyi 276000, P. R. China
| |
Collapse
|
34
|
Zhang H, Shen Y, Kim IM, Weintraub NL, Tang Y. The Impaired Bioenergetics of Diabetic Cardiac Microvascular Endothelial Cells. Front Endocrinol (Lausanne) 2021; 12:642857. [PMID: 34054724 PMCID: PMC8160466 DOI: 10.3389/fendo.2021.642857] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/06/2021] [Indexed: 01/22/2023] Open
Abstract
Diabetes causes hyperglycemia, which can create a stressful environment for cardiac microvascular endothelial cells (CMECs). To investigate the impact of diabetes on the cellular metabolism of CMECs, we assessed glycolysis by quantifying the extracellular acidification rate (ECAR), and mitochondrial oxidative phosphorylation (OXPHOS) by measuring cellular oxygen consumption rate (OCR), in isolated CMECs from wild-type (WT) hearts and diabetic hearts (db/db) using an extracellular flux analyzer. Diabetic CMECs exhibited a higher level of intracellular reactive oxygen species (ROS), and significantly reduced glycolytic reserve and non-glycolytic acidification, as compared to WT CMECs. In addition, OCR assay showed that diabetic CMECs had increased maximal respiration, and significantly reduced non-mitochondrial oxygen consumption and proton leak. Quantitative PCR (qPCR) showed no difference in copy number of mitochondrial DNA (mtDNA) between diabetic and WT CMECs. In addition, gene expression profiling analysis showed an overall decrease in the expression of essential genes related to β-oxidation (Sirt1, Acox1, Acox3, Hadha, and Hadhb), tricarboxylic acid cycle (TCA) (Idh-3a and Ogdh), and electron transport chain (ETC) (Sdhd and Uqcrq) in diabetic CMECs compared to WT CMECs. Western blot confirmed that the protein expression of Hadha, Acox1, and Uqcrq was decreased in diabetic CMECs. Although lectin staining demonstrated no significant difference in capillary density between the hearts of WT mice and db/db mice, diabetic CMECs showed a lower percentage of cell proliferation by Ki67 staining, and a higher percentage of cellular apoptosis by TUNEL staining, compared with WT CMECs. In conclusion, excessive ROS caused by hyperglycemia is associated with impaired glycolysis and mitochondrial function in diabetic CMECs, which in turn may reduce proliferation and promote CMEC apoptosis.
Collapse
Affiliation(s)
- Haitao Zhang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yan Shen
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Il-man Kim
- Anatomy, Cell Biology & Physiology, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Neal L. Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- *Correspondence: Yaoliang Tang,
| |
Collapse
|
35
|
Xiao M, Tang Y, Wang S, Wang J, Wang J, Guo Y, Zhang J, Gu J. The Role of Fibroblast Growth Factor 21 in Diabetic Cardiovascular Complications and Related Epigenetic Mechanisms. Front Endocrinol (Lausanne) 2021; 12:598008. [PMID: 34349728 PMCID: PMC8326758 DOI: 10.3389/fendo.2021.598008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 06/17/2021] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor 21 (FGF21), is an emerging metabolic regulator mediates multiple beneficial effects in the treatment of metabolic disorders and related complications. Recent studies showed that FGF21 acts as an important inhibitor in the onset and progression of cardiovascular complications of diabetes mellitus (DM). Furthermore, evidences discussed so far demonstrate that epigenetic modifications exert a crucial role in the initiation and development of DM-related cardiovascular complications. Thus, epigenetic modifications may involve in the function of FGF21 on DM-induced cardiovascular complications. Therefore, this review mainly interprets and delineates the recent advances of role of FGF21 in DM cardiovascular complications. Then, the possible changes of epigenetics related to the role of FGF21 on DM-induced cardiovascular complications are discussed. Thus, this article not only implies deeper understanding of the pathological mechanism of DM-related cardiovascular complications, but also provides the possible novel therapeutic strategy for DM-induced cardiovascular complications by targeting FGF21 and related epigenetic mechanism.
Collapse
Affiliation(s)
- Mengjie Xiao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Shudong Wang
- Department of Cardiology at the First Hospital of Jilin University, Changchun, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingjing Zhang
- Department of Cardiology at the First Hospital of China Medical University, and Department of Cardiology at the People’s Hospital of Liaoning Province, Shenyang, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Junlian Gu,
| |
Collapse
|
36
|
MicroRNA-34a: the bad guy in age-related vascular diseases. Cell Mol Life Sci 2021; 78:7355-7378. [PMID: 34698884 PMCID: PMC8629897 DOI: 10.1007/s00018-021-03979-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
The age-related vasculature alteration is the prominent risk factor for vascular diseases (VD), namely, atherosclerosis, abdominal aortic aneurysm, vascular calcification (VC) and pulmonary arterial hypertension (PAH). The chronic sterile low-grade inflammation state, alias inflammaging, characterizes elderly people and participates in VD development. MicroRNA34-a (miR-34a) is emerging as an important mediator of inflammaging and VD. miR-34a increases with aging in vessels and induces senescence and the acquisition of the senescence-associated secretory phenotype (SASP) in vascular smooth muscle (VSMCs) and endothelial (ECs) cells. Similarly, other VD risk factors, including dyslipidemia, hyperglycemia and hypertension, modify miR-34a expression to promote vascular senescence and inflammation. miR-34a upregulation causes endothelial dysfunction by affecting ECs nitric oxide bioavailability, adhesion molecules expression and inflammatory cells recruitment. miR-34a-induced senescence facilitates VSMCs osteoblastic switch and VC development in hyperphosphatemia conditions. Conversely, atherogenic and hypoxic stimuli downregulate miR-34a levels and promote VSMCs proliferation and migration during atherosclerosis and PAH. MiR34a genetic ablation or miR-34a inhibition by anti-miR-34a molecules in different experimental models of VD reduce vascular inflammation, senescence and apoptosis through sirtuin 1 Notch1, and B-cell lymphoma 2 modulation. Notably, pleiotropic drugs, like statins, liraglutide and metformin, affect miR-34a expression. Finally, human studies report that miR-34a levels associate to atherosclerosis and diabetes and correlate with inflammatory factors during aging. Herein, we comprehensively review the current knowledge about miR-34a-dependent molecular and cellular mechanisms activated by VD risk factors and highlight the diagnostic and therapeutic potential of modulating its expression in order to reduce inflammaging and VD burn and extend healthy lifespan.
Collapse
|
37
|
Zoja C, Xinaris C, Macconi D. Diabetic Nephropathy: Novel Molecular Mechanisms and Therapeutic Targets. Front Pharmacol 2020; 11:586892. [PMID: 33519447 PMCID: PMC7845653 DOI: 10.3389/fphar.2020.586892] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetic nephropathy (DN) is one of the major microvascular complications of diabetes mellitus and the leading cause of end-stage kidney disease. The standard treatments for diabetic patients are glucose and blood pressure control, lipid lowering, and renin-angiotensin system blockade; however, these therapeutic approaches can provide only partial renoprotection if started late in the course of the disease. One major limitation in developing efficient therapies for DN is the complex pathobiology of the diabetic kidney, which undergoes a set of profound structural, metabolic and functional changes. Despite these difficulties, experimental models of diabetes have revealed promising therapeutic targets by identifying pathways that modulate key functions of podocytes and glomerular endothelial cells. In this review we will describe recent advances in the field, analyze key molecular pathways that contribute to the pathogenesis of the disease, and discuss how they could be modulated to prevent or reverse DN.
Collapse
Affiliation(s)
- Carlamaria Zoja
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Christodoulos Xinaris
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy.,University of Nicosia Medical School, Nicosia, Cyprus
| | - Daniela Macconi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| |
Collapse
|
38
|
Ibrahim AA, Wahby AA, Ashmawy I, Saleh RM, Soliman H. Association of Exosomal miR-34a with Markers of Dyslipidemia and Endothelial Dysfunction in Children and Adolescents with T1DM. J Clin Res Pediatr Endocrinol 2020; 12:401-409. [PMID: 32654473 PMCID: PMC7711647 DOI: 10.4274/jcrpe.galenos.2020.2020.0134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Dyslipidemia and endothelial dysfunction are common disorders and major causative factors for atherosclerosis in patients with type 1 diabetes mellitus (T1DM). However, their pathophysiology in young patients with T1DM is still under evaluated. We aimed, for the first time, to assess the expression of exosomal micro-RNA 34a (miR-34a) in serum of children and adolescents with T1DM and correlate this expression with markers of dyslipidemia and endothelial dysfunction. METHODS The study included 120 T1DM patients and 100 control subjects. Assessment of miR-34a was performed using quantitative real-time polymerase chain reaction. Lipid profile was assessed on an automated analyzer and serum endoglin and intracellular adhesion molecule (ICAM) concentrations were measured using immunometric methods. RESULTS Relative expression of miR-34a and serum endoglin and ICAM concentrations were higher in patients than controls (p=0.001) and in patients with dyslipidemia (42 patients) compared to patients without dyslipidemia (78 patients) (p=0.01). Linear regression analysis revealed a strong independent association between exosomal miR-34a expression and total cholesterol, low-density lipoprotein, serum endoglin and serum ICAM after adjustment for other cofactors. The utility of miR-34a as an indicator for associated dyslipidemia was tested using receiver operator characteristic curve analysis which revealed area under the curve: 0.73 with confidence interval: 0.63-0.83 and p=0.001. CONCLUSION This was the first study to show the altered expression of exosomal miR-34a among children and adolescents with T1DM. Moreover, association of miR-34a with markers of dyslipidemia and endothelial dysfunction was identified, suggesting that it could play a role in regulation of lipid metabolism and endothelial function in T1DM.
Collapse
Affiliation(s)
- Alshaymaa A. Ibrahim
- National Research Centre, Department of Clinical and Chemical Pathology, Cairo, Egypt,* Address for Correspondence: National Research Centre, Department of Clinical and Chemical Pathology, Cairo, Egypt Phone: +01020500501 E-mail:
| | - Aliaa A. Wahby
- National Research Centre, Department of Clinical and Chemical Pathology, Cairo, Egypt
| | - Ingy Ashmawy
- National Research Centre, Department of Clinical and Chemical Pathology, Cairo, Egypt
| | - Rehan M. Saleh
- National Research Centre, Department of Community, Cairo, Egypt
| | - Hend Soliman
- Cairo University Faculty of Medicine, New Children Hospital, Cairo, Egypt
| |
Collapse
|
39
|
Meng T, Qin W, Liu B. SIRT1 Antagonizes Oxidative Stress in Diabetic Vascular Complication. Front Endocrinol (Lausanne) 2020; 11:568861. [PMID: 33304318 PMCID: PMC7701141 DOI: 10.3389/fendo.2020.568861] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/21/2020] [Indexed: 12/17/2022] Open
Abstract
Diabetic mellitus (DM) is a significant public health concern worldwide with an increased incidence of morbidity and mortality, which is particularly due to the diabetic vascular complications. Several pivotal underlying mechanisms are associated with vascular complications, including hyperglycemia, mitochondrial dysfunction, inflammation, and most importantly, oxidative stress. Oxidative stress triggers defective angiogenesis, activates pro-inflammatory pathways and causes long-lasting epigenetic changes to facilitate the development of vascular complications. Therefore, therapeutic interventions targeting oxidative stress are promising to manage diabetic vascular complications. Sirtuin1 (SIRT1), a class III histone deacetylase belonging to the sirtuin family, plays critical roles in regulating metabolism and ageing-related pathological conditions, such as vascular diseases. Growing evidence has indicated that SIRT1 acts as a sensing regulator in response to oxidative stress and attenuates vascular dysfunction via cooperating with adenosine-monophosphate-activated protein kinase (AMPK) to activate antioxidant signals through various downstream effectors, including peroxisome proliferator-activated receptor-gamma co-activator 1 (PGC-1α), forkhead transcription factors (FOXOs), and peroxisome proliferative-activated receptor α (PPARα). In addition, SIRT1 interacts with hydrogen sulfide (H2S), regulates NADPH oxidase, endothelial NO synthase, and mechanistic target of rapamycin (mTOR) to suppress oxidative stress. Furthermore, mRNA expression of sirt1 is affected by microRNAs in DM. In the current review, we summarize recent advances illustrating the importance of SIRT1 in antagonizing oxidative stress. We also discuss whether modulation of SIRT1 can serve as a therapeutic strategy to treat diabetic vascular complications.
Collapse
Affiliation(s)
- Teng Meng
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen, China
| | - Weifeng Qin
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
40
|
Jiang B, Wang R, Lin Z, Ma J, Cui J, Wang M, Liu R, Wu W, Zhang C, Li W, Wang S. Antisense long non‑coding RNA WEE2‑AS1 regulates human vascular endothelial cell viability via cell cycle G2/M transition in arteriosclerosis obliterans. Mol Med Rep 2020; 22:5069-5082. [PMID: 33174040 PMCID: PMC7646961 DOI: 10.3892/mmr.2020.11625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 07/03/2020] [Indexed: 12/19/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) affect atherosclerosis by regulating the physiological and pathological processes of endothelial cells; however, the role of lncRNA WEE2 antisense RNA 1 (WEE2-AS1) in arteriosclerosis obliterans (ASO) is not completely understood. The present study aimed to explore the function of lncRNA WEE2-AS1 in human vascular endothelial cells. The results indicated that lncRNA WEE2-AS1 was significantly elevated in plasma and artery tissue samples of patients with ASO compared with healthy controls. The fluorescence in situ hybridization results suggested that lncRNA WEE2-AS1 was expressed in the cytoplasm and nuclei of primary human umbilical vein endothelial cells (HUVECs). The Cell Counting Kit-8 assay results suggested that lncRNA WEE2-AS1 knockdown significantly promoted HUVEC viability, whereas lncRNA WEE2-AS1 overexpression inhibited HUVEC viability compared with the negative control groups. Furthermore, analysis of the cell cycle by flow cytometry indicated that lncRNA WEE2-AS1 knockdown significantly decreased the proportion of cells in the G0/G1 phase and significantly increased the proportion of cells in the G2/M phase compared with the negative control group. However, lncRNA WEE2-AS1 overexpression had no significant effect on cell cycle distribution compared with the negative control group. The western blotting results indicated that lncRNA WEE2-AS1 knockdown significantly reduced the expression levels of phosphorylated cyclin dependent kinase 1, WEE1 homolog 2 and myelin transcription factor 1, but increased the expression level of cell division cycle 25B compared with the negative control group. lncRNA WEE2-AS1 overexpression displayed the opposite effect on protein expression. Collectively, the present study suggested that lncRNA WEE2-AS1 was significantly upregulated in ASO and may serve a role in regulating human vascular endothelial cell viability. Further investigation into lncRNA WEE2-AS1 may broaden the current understanding of the molecular mechanism underlying ASO, and aid with the identification of specific probes and precise targeted drugs for the diagnosis and treatment of ASO.
Collapse
Affiliation(s)
- Baohong Jiang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Rui Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zefei Lin
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jieyi Ma
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jin Cui
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Mian Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Ruiming Liu
- National‑Local Joint Engineering Laboratory of Vascular Disease Diagnosis and Treatment, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Weibin Wu
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Chunxiang Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Wen Li
- National‑Local Joint Engineering Laboratory of Vascular Disease Diagnosis and Treatment, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Shenming Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
41
|
Raucci A, Vinci MC. miR-34a: A Promising Target for Inflammaging and Age-Related Diseases. Int J Mol Sci 2020; 21:ijms21218293. [PMID: 33167452 PMCID: PMC7663903 DOI: 10.3390/ijms21218293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 01/13/2023] Open
Affiliation(s)
- Angela Raucci
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy
- Correspondence: (A.R.); (M.C.V.); Tel.: +39-02-5800-2802 (A.R.); +39-02-5800-2028 (M.C.V.); Fax: +39-02-5800-2342 (A.R. & M.C.V.)
| | - Maria Cristina Vinci
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy
- Correspondence: (A.R.); (M.C.V.); Tel.: +39-02-5800-2802 (A.R.); +39-02-5800-2028 (M.C.V.); Fax: +39-02-5800-2342 (A.R. & M.C.V.)
| |
Collapse
|
42
|
Hosen MR, Goody PR, Zietzer A, Nickenig G, Jansen F. MicroRNAs As Master Regulators of Atherosclerosis: From Pathogenesis to Novel Therapeutic Options. Antioxid Redox Signal 2020; 33:621-644. [PMID: 32408755 DOI: 10.1089/ars.2020.8107] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Cardiovascular disease (CVD) remains the major cause of morbidity and mortality worldwide. Accumulating evidence indicates that atherosclerosis and its sequelae, coronary artery disease, contribute to the majority of cardiovascular deaths. Atherosclerosis is a chronic inflammatory disease of the arteries in which atherosclerotic plaques form within the vessel wall. Epidemiological studies have identified various risk factors for atherosclerosis, such as diabetes, hyperlipidemia, smoking, genetic predisposition, and sedentary lifestyle. Recent Advances: Through the advancement of genetic manipulation techniques and their use in cardiovascular biology, it was shown that small RNAs, especially microRNAs (miRNAs), are dynamic regulators of disease pathogenesis. They are considered to be central during the regulation of gene expression through numerous mechanisms and provide a means to develop biomarkers and therapeutic tools for the diagnosis and therapy of atherosclerosis. Circulating miRNAs encapsulated within membrane-surrounded vesicles, which originate from diverse subcellular compartments, are now emerging as novel regulators of intercellular communication. The miRNAs, in both freely circulating and vesicle-bound forms, represent a valuable tool for diagnosing and monitoring CVD, recently termed as "liquid biopsy." Critical Issues: However, despite the recent advancements in miRNA-based diagnostics and therapeutics, understanding how miRNAs can regulate atherosclerosis is still crucial to achieving an effective intervention and reducing the disease burden. Future Directions: We provide a landscape of the current developmental progression of RNA therapeutics as a holistic approach for treating CVD in different animal models and clinical trials. Future interrogations are warranted for the development of miRNA-based therapeutics to overcome challenges for the treatment of the disease.
Collapse
Affiliation(s)
- Mohammed Rabiul Hosen
- Department of Internal Medicine II, Molecular Cardiology, Heart Center Bonn, Rheinische Friedrich-Wilhelms University Bonn, Bonn, Germany
| | - Philip Roger Goody
- Department of Internal Medicine II, Molecular Cardiology, Heart Center Bonn, Rheinische Friedrich-Wilhelms University Bonn, Bonn, Germany
| | - Andreas Zietzer
- Department of Internal Medicine II, Molecular Cardiology, Heart Center Bonn, Rheinische Friedrich-Wilhelms University Bonn, Bonn, Germany
| | - Georg Nickenig
- Department of Internal Medicine II, Molecular Cardiology, Heart Center Bonn, Rheinische Friedrich-Wilhelms University Bonn, Bonn, Germany
| | - Felix Jansen
- Department of Internal Medicine II, Molecular Cardiology, Heart Center Bonn, Rheinische Friedrich-Wilhelms University Bonn, Bonn, Germany
| |
Collapse
|
43
|
Climent M, Viggiani G, Chen YW, Coulis G, Castaldi A. MicroRNA and ROS Crosstalk in Cardiac and Pulmonary Diseases. Int J Mol Sci 2020; 21:ijms21124370. [PMID: 32575472 PMCID: PMC7352701 DOI: 10.3390/ijms21124370] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/14/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) affect many cellular functions and the proper redox balance between ROS and antioxidants contributes substantially to the physiological welfare of the cell. During pathological conditions, an altered redox equilibrium leads to increased production of ROS that in turn may cause oxidative damage. MicroRNAs (miRNAs) regulate gene expression at the post-transcriptional level contributing to all major cellular processes, including oxidative stress and cell death. Several miRNAs are expressed in response to ROS to mediate oxidative stress. Conversely, oxidative stress may lead to the upregulation of miRNAs that control mechanisms to buffer the damage induced by ROS. This review focuses on the complex crosstalk between miRNAs and ROS in diseases of the cardiac (i.e., cardiac hypertrophy, heart failure, myocardial infarction, ischemia/reperfusion injury, diabetic cardiomyopathy) and pulmonary (i.e., idiopathic pulmonary fibrosis, acute lung injury/acute respiratory distress syndrome, asthma, chronic obstructive pulmonary disease, lung cancer) compartments. Of note, miR-34a, miR-144, miR-421, miR-129, miR-181c, miR-16, miR-31, miR-155, miR-21, and miR-1/206 were found to play a role during oxidative stress in both heart and lung pathologies. This review comprehensively summarizes current knowledge in the field.
Collapse
Affiliation(s)
- Montserrat Climent
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20089 Rozzano, MI, Italy;
| | - Giacomo Viggiani
- Department of Biomedical Sciences, Humanitas University, 20090 Pieve Emanuele, MI, Italy;
| | - Ya-Wen Chen
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Gerald Coulis
- Department of Physiology and Biophysics, and Institute for Immunology, University of California Irvine, Irvine, CA 92697, USA;
| | - Alessandra Castaldi
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Correspondence:
| |
Collapse
|
44
|
Sirtuins family as a target in endothelial cell dysfunction: implications for vascular ageing. Biogerontology 2020; 21:495-516. [PMID: 32285331 DOI: 10.1007/s10522-020-09873-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/20/2020] [Indexed: 12/13/2022]
Abstract
The vascular endothelium is a protective barrier between the bloodstream and the vasculature that may be disrupted by different factors such as the presence of diseased states. Diseases like diabetes and obesity pose a great risk toward endothelial cell inflammation and oxidative stress, leading to endothelial cell dysfunction and thereby cardiovascular complications such as atherosclerosis. Sirtuins are NAD+-dependent histone deacetylases that are implicated in the pathophysiology of cardiovascular diseases, and they have been identified to be important regulators of endothelial cell function. A handful of recent studies suggest that disbalance in the regulation of endothelial sirtuins, mainly sirtuin 1 (SIRT1), contributes to endothelial cell dysfunction. Herein, we summarize how SIRT1 and other sirtuins may contribute to endothelial cell function and how presence of diseased conditions may alter their expressions to cause endothelial dysfunction. Moreover, we discuss how the beneficial effects of exercise on the endothelium are dependent on SIRT1. These mainly include regulation of signaling pathways related to endothelial nitric oxide synthase phosphorylation and nitric oxide production, mitochondrial biogenesis and mitochondria-mediated apoptotic pathways, oxidative stress and inflammatory pathways. Sirtuins as modulators of the adverse conditions in the endothelium hold a promising therapeutic potential for health conditions related to endothelial dysfunction and vascular ageing.
Collapse
|
45
|
Zhang J, Li Y, Wang B, Luo Y, Shi J, Zhao B. The p66shc-mediated Regulation of Hepatocyte Senescence Influences Hepatic Steatosis in Nonalcoholic Fatty Liver Disease. Med Sci Monit 2020; 26:e921887. [PMID: 32191680 PMCID: PMC7104657 DOI: 10.12659/msm.921887] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Recent studies have suggested that hepatocyte senescence could contribute to hepatic steatosis and its progression in nonalcoholic fatty liver disease (NAFLD). However, the underlying mechanism causing hepatocyte senescence in this pathological condition is still unclear. A thorough understanding of the mechanism could provide a new target for therapeutic intervention. The purpose of this study was to investigate the role of p66shc in hepatocyte senescence and hepatocyte damage in NAFLD progression. Material/Methods We examined the expression levels of hepatic p66shc and senescence markers in rats and humans with NAFLD, and we assessed the effect of p66shc knockdown or overexpression on senescence and steatosis in human liver cells. Results In this study, we showed that increased hepatic p66shc expression was consistent with upregulated expression of the following senescence markers in NAFLD rats: heterochromatin protein-1-beta (HP1β), p16, p21, and p53. Furthermore, senescence and steatosis could be induced in hepatoblastoma cell line (HepG2) cells when cells were stimulated with a low concentration of H2O2, and this effect was significantly alleviated by knockdown of p66shc. However, overexpression of p66shc could promote senescence and steatosis in L02 cells. Finally, increased hepatic p66shc protein levels correlated with enhanced expression of the senescence marker p21 and mirrored the degree of disease severity in NAFLD patients. Conclusions Our findings indicated that the increase in hepatocyte senescence and steatosis in NAFLD may be caused by the upregulation of p66shc expression, implying that strategies for p66shc-mediated regulation of hepatocyte senescence may provide new therapeutic tools for NAFLD.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Yanpeng Li
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Bingyuan Wang
- Department of Elderly Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yan Luo
- Center for Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China (mainland)
| | - Junping Shi
- Department of Liver Diseases, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China (mainland)
| | - Baiyun Zhao
- Drug Clinical Trial Institution, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
46
|
Wang D, Wang T, Wang R, Zhang X, Wang L, Xiang Z, Zhuang L, Shen S, Wang H, Gao Q, Wang Y. Suppression of p66Shc prevents hyperandrogenism-induced ovarian oxidative stress and fibrosis. J Transl Med 2020; 18:84. [PMID: 32066482 PMCID: PMC7027222 DOI: 10.1186/s12967-020-02249-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 01/28/2020] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Rats with hyperandrogen-induced polycystic ovary syndrome (PCOS) have been shown to develop ovarian oxidative stress (OS) and fibrosis. The Sirt1 agonist, resveratrol, can reduce OS through inhibiting p66Shc in other models of OS. METHODS We created a rat PCOS model with increased OS levels following treatment with one of the two androgens, dehydroepiandrosterone (DHEA) and dihydrotestosterone (DHT). The PCOS related features were determined by measurement of malondialdehyde (MDA) and superoxide dismutase (SOD) levels or by examining the reactive oxygen species (ROS) levels using the DCF-DA probe. The potential mechanisms by which p66Shc/Sirt1 mediates ovarian fibrosis were explored by western blotting, quantitative reverse transcription-PCR, immunofluorescence staining, and immunohistochemistry. RESULTS Hyperandrogen dramatically augmented OS and activation of fibrotic factors in the ovary. Our data demonstrated that treatment with resveratrol enhanced Sirt1 and decreased ovarian OS as well as inhibited phosphorylation of p66Shc both in vivo and in vitro. The treatment suppressed fibrotic factor activation and improved ovarian morphology. Lentivirus- or siRNA-mediated p66Shc knockdown resulted in a dramatic enhancement of Sirt1 expression, down-regulation of ROS and suppression of fibrotic factors in granulosa cells. Moreover, p66Shc overexpression markedly increased the expression of fibrotic factors. Additionally, silencing Sirt1 induced a dramatic increase in p66Shc and enhanced activation of fibrotic factors. CONCLUSIONS p66Shc may be a direct target of Sirt1 for inducing ROS and thus promoting fibrosis. Further exploration of the mechanisms of p66Shc in both fibrosis and OS may provide novel therapeutic strategies that will facilitate the improvement in PCOS symptoms and reproductive functions.
Collapse
Affiliation(s)
- Daojuan Wang
- State Key Laboratory of Analytacal Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Tingyu Wang
- State Key Laboratory of Analytacal Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Rong Wang
- State Key Laboratory of Analytacal Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Xinlin Zhang
- Department of Cardiology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, 321 Zhongshan Road, 210008, Nanjing, Jiangsu Province, China
| | - Lei Wang
- State Key Laboratory of Analytacal Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Zou Xiang
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Lingjia Zhuang
- State Key Laboratory of Analytacal Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Shanmei Shen
- Department of Endocrinology, The Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Hongwei Wang
- State Key Laboratory of Analytacal Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Qian Gao
- State Key Laboratory of Analytacal Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Yong Wang
- State Key Laboratory of Analytacal Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
47
|
Piao S, Nagar H, Kim S, Lee I, Choi SJ, Kim T, Jeon BH, Kim CS. CRIF1 deficiency induced mitophagy via p66shc-regulated ROS in endothelial cells. Biochem Biophys Res Commun 2020; 522:869-875. [DOI: 10.1016/j.bbrc.2019.11.109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/17/2019] [Indexed: 12/15/2022]
|
48
|
Grieco GE, Brusco N, Licata G, Nigi L, Formichi C, Dotta F, Sebastiani G. Targeting microRNAs as a Therapeutic Strategy to Reduce Oxidative Stress in Diabetes. Int J Mol Sci 2019; 20:ijms20246358. [PMID: 31861156 PMCID: PMC6940935 DOI: 10.3390/ijms20246358] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/09/2019] [Accepted: 12/15/2019] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is a group of heterogeneous metabolic disorders characterized by chronic hyperglycaemia as a consequence of pancreatic β cell loss and/or dysfunction, also caused by oxidative stress. The molecular mechanisms involved inβ cell dysfunction and in response to oxidative stress are also regulated by microRNAs (miRNAs). miRNAs are a class of negative gene regulators, which modulate pathologic mechanisms occurring in diabetes and its complications. Although several pharmacological therapies specifically targeting miRNAs have already been developed and brought to the clinic, most previous miRNA-based drug delivery methods were unable to target a specific miRNA in a single cell type or tissue, leading to important off-target effects. In order to overcome these issues, aptamers and nanoparticles have been described as non-cytotoxic vehicles for miRNA-based drug delivery. These approaches could represent an innovative way to specifically target and modulate miRNAs involved in oxidative stress in diabetes and its complications. Therefore, the aims of this review are: (i) to report the role of miRNAs involved in oxidative stress in diabetes as promising therapeutic targets; (ii) to shed light onto the new delivery strategies developed to modulate the expression of miRNAs in diseases.
Collapse
Affiliation(s)
- Giuseppina Emanuela Grieco
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, V.le Bracci, 16, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (L.N.); (C.F.); (G.S.)
- Fondazione Umberto Di Mario ONLUS c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Noemi Brusco
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, V.le Bracci, 16, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (L.N.); (C.F.); (G.S.)
- Fondazione Umberto Di Mario ONLUS c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Giada Licata
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, V.le Bracci, 16, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (L.N.); (C.F.); (G.S.)
- Fondazione Umberto Di Mario ONLUS c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, V.le Bracci, 16, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (L.N.); (C.F.); (G.S.)
- Fondazione Umberto Di Mario ONLUS c/o Toscana Life Sciences, 53100 Siena, Italy
- UO Diabetologia, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| | - Caterina Formichi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, V.le Bracci, 16, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (L.N.); (C.F.); (G.S.)
- Fondazione Umberto Di Mario ONLUS c/o Toscana Life Sciences, 53100 Siena, Italy
- UO Diabetologia, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, V.le Bracci, 16, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (L.N.); (C.F.); (G.S.)
- Fondazione Umberto Di Mario ONLUS c/o Toscana Life Sciences, 53100 Siena, Italy
- UO Diabetologia, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
- Correspondence: ; Tel.: +39-0577-586269
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, V.le Bracci, 16, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (L.N.); (C.F.); (G.S.)
- Fondazione Umberto Di Mario ONLUS c/o Toscana Life Sciences, 53100 Siena, Italy
| |
Collapse
|
49
|
Qadir MMF, Klein D, Álvarez-Cubela S, Domínguez-Bendala J, Pastori RL. The Role of MicroRNAs in Diabetes-Related Oxidative Stress. Int J Mol Sci 2019; 20:E5423. [PMID: 31683538 PMCID: PMC6862492 DOI: 10.3390/ijms20215423] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022] Open
Abstract
Cellular stress, combined with dysfunctional, inadequate mitochondrial phosphorylation, produces an excessive amount of reactive oxygen species (ROS) and an increased level of ROS in cells, which leads to oxidation and subsequent cellular damage. Because of its cell damaging action, an association between anomalous ROS production and disease such as Type 1 (T1D) and Type 2 (T2D) diabetes, as well as their complications, has been well established. However, there is a lack of understanding about genome-driven responses to ROS-mediated cellular stress. Over the last decade, multiple studies have suggested a link between oxidative stress and microRNAs (miRNAs). The miRNAs are small non-coding RNAs that mostly suppress expression of the target gene by interaction with its 3'untranslated region (3'UTR). In this paper, we review the recent progress in the field, focusing on the association between miRNAs and oxidative stress during the progression of diabetes.
Collapse
Affiliation(s)
- Mirza Muhammad Fahd Qadir
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Dagmar Klein
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Ricardo Luis Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
50
|
Golshiri K, Ataei Ataabadi E, Portilla Fernandez EC, Jan Danser AH, Roks AJM. The importance of the nitric oxide-cGMP pathway in age-related cardiovascular disease: Focus on phosphodiesterase-1 and soluble guanylate cyclase. Basic Clin Pharmacol Toxicol 2019; 127:67-80. [PMID: 31495057 DOI: 10.1111/bcpt.13319] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 08/29/2019] [Indexed: 12/18/2022]
Abstract
Among ageing-related illnesses, cardiovascular disease (CVD) remains the leading cause of morbidity and mortality causing one-third of all deaths worldwide. Ageing evokes a number of functional, pharmacological and morphological changes in the vasculature, accompanied by a progressive failure of protective and homeostatic mechanisms, resulting in target organ damage. Impaired vasomotor, proliferation, migration, antithrombotic and anti-inflammatory function in both the endothelial and vascular smooth muscle cells are parts of the vascular ageing phenotype. The endothelium regulates these functions by the release of a wide variety of active molecules including endothelium-derived relaxing factors such as nitric oxide, prostacyclin (PGI2 ) and endothelium-derived hyperpolarization (EDH). During ageing, a functional decay of the nitric oxide pathway takes place. Nitric oxide signals to VSMC and other important cell types for vascular homeostasis through the second messenger cyclic guanosine monophosphate (cGMP). Maintenance of proper cGMP levels is an important goal in sustainment of proper vascular function during ageing. For this purpose, different components can be targeted in this signalling system, and among them, phosphodiesterase-1 (PDE1) and soluble guanylate cyclase (sGC) are crucial. This review focuses on the role of PDE1 and sGC in conditions that are relevant for vascular ageing.
Collapse
Affiliation(s)
- Keivan Golshiri
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ehsan Ataei Ataabadi
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eliana C Portilla Fernandez
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Anton J M Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|