1
|
Kawamura J, Yamakuchi M, Ueno K, Hashiguchi T, Okamoto Y. MiR-25-3p regulates pulmonary arteriovenous malformation after Glenn procedure in patients with univentricular heart via the PHLPP2-HIF-1α axis. Sci Rep 2025; 15:4138. [PMID: 39900983 PMCID: PMC11790876 DOI: 10.1038/s41598-025-88840-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/31/2025] [Indexed: 02/05/2025] Open
Abstract
The detailed mechanism of pulmonary arteriovenous malformations after Glenn surgery (G-PAVMs) in cyanotic congenital heart disease (CHD) remains unclear. Microarray in situ hybridization was performed to assess the miRNA (miRNA) profiles of serum from pediatric patients (0-6 years of age) with G-PAVMs and after the Fontan procedure without G-PAVMs. In addition, we investigated the tube formation, migration, and proliferation of human lung microvascular endothelial cells (HMVEC-L) transfected with miR-25-3p mimic, miR-25-3p inhibitor, or PHLPP2 small interfering RNA, and examined HIF-1α/VEGF-A signaling after hypoxic stimulation. Serum miRNAs that showed ≥ 2-fold higher levels in patients with G-PAVMs than in other patients were selected. MiR-25-3p was significantly upregulated in the pulmonary artery sera of the post-Glenn group than in the post-Fontan group. We identified PHLPP2 as a direct target of miR-25-3p. PHLPP2 expression was significantly decreased in HMVEC-L transfected with miR-25-3p mimic compared to the control cells. HIF-1α and VEGF-A expression levels were increased in HMVEC-L transfected with miR-25-3p mimic compared to the control cells in a PHLPP2/Akt/mTOR signaling-dependent manner after hypoxic stimulation. MiR-25-3p promoted HMVEC-L angiogenesis, proliferation, and migration under hypoxic conditions. MiR-25-3p in the pulmonary arteries may contribute to G-PAVM development.
Collapse
Affiliation(s)
- Junpei Kawamura
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Munekazu Yamakuchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, 890-8544, Japan.
| | - Kentaro Ueno
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Teruto Hashiguchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, 890-8544, Japan
| | - Yasuhiro Okamoto
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
2
|
Huang S, Zhou Y, Zhang Y, Liu N, Liu J, Liu L, Fan C. Advances in MicroRNA Therapy for Heart Failure: Clinical Trials, Preclinical Studies, and Controversies. Cardiovasc Drugs Ther 2025; 39:221-232. [PMID: 37505309 DOI: 10.1007/s10557-023-07492-7] [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] [Accepted: 07/17/2023] [Indexed: 07/29/2023]
Abstract
Heart failure (HF) is a rapidly growing public health issue with more than 37.7 million patients worldwide and an annual healthcare cost of $108 billion. However, HF-related drugs have not changed significantly for decades, and it is essential to find biological drugs to provide better treatment for HF patients. MicroRNAs (miRNAs) are non-coding RNAs (ncRNAs) with a length of approximately 21 nucleotides and play an important role in the onset and progression of cardiovascular diseases. Increasing studies have shown that miRNAs are widely involved in the pathophysiology of HF, and the regulation of miRNAs has promising therapeutic effects. Among them, there is great interest in miRNA-132, since the encouraging success of anti-miRNA-132 therapy in a phase 1b clinical trial in 2020. However, it is worth noting that the multi-target effect of miRNA may produce side effects such as thrombocytopenia, revascularization dysfunction, severe immune response, and even death. Advances in drug delivery modalities, delivery vehicles, chemical modifications, and plant-derived miRNAs are expected to address safety concerns and further improve miRNA therapy. Here, we reviewed the preclinical studies and clinical trials of HF-related miRNAs (especially miRNA-132) in the past 5 years and summarized the controversies of miRNA therapy.
Collapse
Affiliation(s)
- Shengyuan Huang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Yong Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yiru Zhang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ningyuan Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiachen Liu
- Xiangya Medical College of Central South University, Changsha, China
| | - Liming Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China.
| |
Collapse
|
3
|
Song R, Zhang L. MicroRNAs and therapeutic potentials in acute and chronic cardiac disease. Drug Discov Today 2024; 29:104179. [PMID: 39276921 DOI: 10.1016/j.drudis.2024.104179] [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: 06/18/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
microRNAs (miRNAs) are small regulatory RNAs implicated in various cardiac disorders. In this review, the role of miRNAs is discussed in relation to acute myocardial infarction and chronic heart failure. In both settings, miRNAs are altered, contributing to injury and adverse remodeling. Notably, miRNA profiles differ between acute ischemic injury and progressive heart failure. Owing to miRNA variabilities between disease stages and delivery difficulties, translation of animal studies to the clinic remains challenging. The identification of distinct miRNA signatures could lead to the development of miRNA therapies tailored to different disease stages. Here, we summarize the current understanding of miRNAs in acute and chronic cardiac diseases, identify knowledge gaps and discuss progress in developing miRNA-based treatment strategies.
Collapse
Affiliation(s)
- Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
4
|
Kepreotis SV, Oh JG, Park M, Yoo J, Lee C, Mercola M, Hajjar RJ, Jeong D. Inhibition of miR-25 ameliorates cardiac and skeletal muscle dysfunction in aged mdx/utrn haploinsufficient (+/-) mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102174. [PMID: 38584818 PMCID: PMC10998245 DOI: 10.1016/j.omtn.2024.102174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/14/2024] [Indexed: 04/09/2024]
Abstract
Dystrophic cardiomyopathy is a significant feature of Duchenne muscular dystrophy (DMD). Increased cardiomyocyte cytosolic calcium (Ca2+) and interstitial fibrosis are major pathophysiological hallmarks that ultimately result in cardiac dysfunction. MicroRNA-25 (miR-25) has been identified as a suppressor of both sarcoplasmic reticulum calcium ATPase 2a (SERCA2a) and mothers against decapentaplegic homolog-7 (Smad7) proteins. In this study, we created a gene transfer using an miR-25 tough decoy (TuD) RNA inhibitor delivered via recombinant adeno-associated virus serotype 9 (AAV9) to evaluate the effect of miR-25 inhibition on cardiac and skeletal muscle function in aged dystrophin/utrophin haploinsufficient mice mdx/utrn (+/-), a validated transgenic murine model of DMD. We found that the intravenous delivery of AAV9 miR-25 TuD resulted in strong and stable inhibition of cardiac miR-25 levels, together with the restoration of SERCA2a and Smad7 expression. This was associated with the amelioration of cardiomyocyte interstitial fibrosis as well as recovered cardiac function. Furthermore, the direct quadricep intramuscular injection of AAV9 miR-25 TuD significantly restored skeletal muscle Smad7 expression, reduced tissue fibrosis, and enhanced skeletal muscle performance in mdx/utrn (+/-) mice. These results imply that miR-25 TuD gene transfer may be a novel therapeutic approach to restore cardiomyocyte Ca2+ homeostasis and abrogate tissue fibrosis in DMD.
Collapse
Affiliation(s)
- Sacha V. Kepreotis
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Jae Gyun Oh
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Mina Park
- Department of Medicinal and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
| | - Jimeen Yoo
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Cholong Lee
- Department of Medicinal and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
| | - Mark Mercola
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Roger J. Hajjar
- Mass General Brigham Gene and Cell Therapy Institute, Boston, MA, USA
| | - Dongtak Jeong
- Department of Medicinal and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| |
Collapse
|
5
|
Wójtowicz A, Molcan T, Lukasik K, Żebrowska E, Pawlina-Tyszko K, Gurgul A, Szmatoła T, Bugno-Poniewierska M, Ferreira-Dias G, Skarzynski DJ, Szóstek-Mioduchowska A. The potential role of miRNAs and regulation of their expression in the development of mare endometrial fibrosis. Sci Rep 2023; 13:15938. [PMID: 37743390 PMCID: PMC10518347 DOI: 10.1038/s41598-023-42149-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023] Open
Abstract
Mare endometrial fibrosis (endometrosis), is one of the main causes of equine infertility. Despite the high prevalence, both ethology, pathogenesis and the nature of its progression remain poorly understood. Recent studies have shown that microRNAs (miRNAs) are important regulators in multiple cellular processes and functions under physiological and pathological circumstances. In this article, we reported changes in miRNA expression at different stages of endometrosis and the effect of transforming growth factor (TGF)-β1 on the expression of the most dysregulated miRNAs. We identified 1, 26, and 5 differentially expressed miRNAs (DEmiRs), in categories IIA (mild fibrosis), IIB (moderate fibrosis), and III (severe fibrosis) groups compared to category I (no fibrosis) endometria group, respectively (Padjusted < 0.05, log2FC ≥ 1.0/log2FC ≤ - 1.0). This study indicated the potential involvement of miRNAs in the regulation of the process associated to the development and progression of endometrosis. The functional enrichment analysis revealed, that DEmiRs target genes involved in the mitogen-activated protein kinases, Hippo, and phosphoinositide-3-kinase (PI3K)-Akt signalling pathways, focal adhesion, and extracellular matrix-receptor interaction. Moreover, we demonstrated that the most potent profibrotic cytokine-TGF-β1-downregulated novel-eca-miR-42 (P < 0.05) expression in fibroblasts derived from endometria at early-stage endometrosis (category IIA).
Collapse
Affiliation(s)
- Anna Wójtowicz
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Tomasz Molcan
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Karolina Lukasik
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Ewelina Żebrowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Klaudia Pawlina-Tyszko
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Cracow, Poland
| | - Artur Gurgul
- Department of Animal Reproduction, Anatomy and Genomics, The University of Agriculture in Krakow, Cracow, Poland
| | - Tomasz Szmatoła
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Cracow, Poland
- Center for Experimental and Innovative Medicine, University of Agriculture in Krakow, Cracow, Poland
| | - Monika Bugno-Poniewierska
- Department of Animal Reproduction, Anatomy and Genomics, The University of Agriculture in Krakow, Cracow, Poland
| | - Graca Ferreira-Dias
- Faculty of Veterinary Medicine, CIISA - Center for Interdisciplinary Research in Animal Health, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Dariusz J Skarzynski
- Department of Reproduction and Clinic of Farm Animals, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Anna Szóstek-Mioduchowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| |
Collapse
|
6
|
Kabłak-Ziembicka A, Badacz R, Okarski M, Wawak M, Przewłocki T, Podolec J. Cardiac microRNAs: diagnostic and therapeutic potential. Arch Med Sci 2023; 19:1360-1381. [PMID: 37732050 PMCID: PMC10507763 DOI: 10.5114/aoms/169775] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/18/2023] [Indexed: 09/22/2023] Open
Abstract
MicroRNAs are small non-coding post-translational biomolecules which, when expressed, modify their target genes. It is estimated that microRNAs regulate production of approximately 60% of all human proteins and enzymes that are responsible for major physiological processes. In cardiovascular disease pathophysiology, there are several cells that produce microRNAs, including endothelial cells, vascular smooth muscle cells, macrophages, platelets, and cardiomyocytes. There is a constant crosstalk between microRNAs derived from various cell sources. Atherosclerosis initiation and progression are driven by many pro-inflammatory and pro-thrombotic microRNAs. Atherosclerotic plaque rupture is the leading cause of cardiovascular death resulting from acute coronary syndrome (ACS) and leads to cardiac remodeling and fibrosis following ACS. MicroRNAs are powerful modulators of plaque progression and transformation into a vulnerable state, which can eventually lead to plaque rupture. There is a growing body of evidence which demonstrates that following ACS, microRNAs might inhibit fibroblast proliferation and scarring, as well as harmful apoptosis of cardiomyocytes, and stimulate fibroblast reprogramming into induced cardiac progenitor cells. In this review, we focus on the role of cardiomyocyte-derived and cardiac fibroblast-derived microRNAs that are involved in the regulation of genes associated with cardiomyocyte and fibroblast function and in atherosclerosis-related cardiac ischemia. Understanding their mechanisms may lead to the development of microRNA cocktails that can potentially be used in regenerative cardiology.
Collapse
Affiliation(s)
- Anna Kabłak-Ziembicka
- Department of Interventional Cardiology, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- Noninvasive Cardiovascular Laboratory, the John Paul II Hospital, Krakow, Poland
| | - Rafał Badacz
- Department of Interventional Cardiology, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- Department of Interventional Cardiology, the John Paul II Hospital, Krakow, Poland
| | - Michał Okarski
- Student Scientific Group of Modern Cardiac Therapy at the Department of Interventional Cardiology, Jagiellonian University Medical College, Krakow, Poland
| | - Magdalena Wawak
- Department of Interventional Cardiology, the John Paul II Hospital, Krakow, Poland
| | - Tadeusz Przewłocki
- Noninvasive Cardiovascular Laboratory, the John Paul II Hospital, Krakow, Poland
- Department of Cardiac and Vascular Diseases Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
| | - Jakub Podolec
- Department of Interventional Cardiology, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- Department of Interventional Cardiology, the John Paul II Hospital, Krakow, Poland
| |
Collapse
|
7
|
Lee C, Cho S, Jeong D. Inhibition of miR-25 Ameliorates Cardiac Dysfunction and Fibrosis by Restoring Krüppel-like Factor 4 Expression. Int J Mol Sci 2023; 24:12434. [PMID: 37569807 PMCID: PMC10418969 DOI: 10.3390/ijms241512434] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Cardiac hypertrophy is an adaptive response to various pathological insults, including hypertension. However, sustained hypertrophy can cause impaired calcium regulation, cardiac dysfunction, and remodeling, accompanied by cardiac fibrosis. Our previous study identified miR-25 as a regulator of SERCA2a, and found that the inhibition of miR-25 improved cardiac function and reduced fibrosis by restoring SERCA2a expression in a murine heart failure model. However, the precise mechanism underlying the reduction in fibrosis following miR-25 inhibition remains unclear. Therefore, we postulate that miR-25 may have additional targets that contribute to regulating cardiac fibrosis. Using in silico analysis, Krüppel-like factor 4 (KLF4) was identified as an additional target of miR-25. Further experiments confirmed that KLF4 was directly targeted by miR-25 and that its expression was reduced by long-term treatment with Angiotensin II, a major hypertrophic inducer. Subsequently, treatment with an miR-25 inhibitor alleviated the cardiac dysfunction, fibrosis, and inflammation induced by Angiotensin II (Ang II). These findings indicate that inhibiting miR-25 not only enhances calcium cycling and cardiac function via SERCA2a restoration but also reduces fibrosis by restoring KLF4 expression. Therefore, targeting miR-25 may be a promising therapeutic strategy for treating hypertensive heart diseases.
Collapse
Affiliation(s)
| | | | - Dongtak Jeong
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea; (C.L.); (S.C.)
| |
Collapse
|
8
|
Kho C. Targeting calcium regulators as therapy for heart failure: focus on the sarcoplasmic reticulum Ca-ATPase pump. Front Cardiovasc Med 2023; 10:1185261. [PMID: 37534277 PMCID: PMC10392702 DOI: 10.3389/fcvm.2023.1185261] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/06/2023] [Indexed: 08/04/2023] Open
Abstract
Impaired myocardial Ca2+ cycling is a critical contributor to the development of heart failure (HF), causing changes in the contractile function and structure remodeling of the heart. Within cardiomyocytes, the regulation of sarcoplasmic reticulum (SR) Ca2+ storage and release is largely dependent on Ca2+ handling proteins, such as the SR Ca2+ ATPase (SERCA2a) pump. During the relaxation phase of the cardiac cycle (diastole), SERCA2a plays a critical role in transporting cytosolic Ca2+ back to the SR, which helps to restore both cytosolic Ca2+ levels to their resting state and SR Ca2+ content for the next contraction. However, decreased SERCA2a expression and/or pump activity are key features in HF. As a result, there is a growing interest in developing therapeutic approaches to target SERCA2a. This review provides an overview of the regulatory mechanisms of the SERCA2a pump and explores potential strategies for SERCA2a-targeted therapy, which are being investigated in both preclinical and clinical studies.
Collapse
Affiliation(s)
- Changwon Kho
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
9
|
Peng S, Wang M, Zhang S, Liu N, Li Q, Kang J, Chen L, Li M, Pang K, Huang J, Lu F, Zhao D, Zhang W. Hydrogen sulfide regulates SERCA2a SUMOylation by S-Sulfhydration of SENP1 to ameliorate cardiac systole-diastole function in diabetic cardiomyopathy. Biomed Pharmacother 2023; 160:114200. [PMID: 36750014 DOI: 10.1016/j.biopha.2022.114200] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/20/2022] [Accepted: 12/30/2022] [Indexed: 02/07/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a serious complication of diabetes mellitus that eventually progresses to heart failure. The sarco(endo)plasmic reticulum calcium ATPase 2a (SERCA2a), an important calcium pump in cardiomyocytes, is closely related to myocardial systolic-diastolic function. In mammalian cells, hydrogen sulfide (H2S), as a second messenger, antioxidant, and sulfurizing agent, is involved in diverse biological processes. Despite the importance of H2S for protection against DCM, the mechanisms remain poorly understood. The aim of the present study was to determine whether H2S regulates intracellular calcium homeostasis by acting on SERCA2a to reduce cardiomyocyte apoptosis during DCM. Db/db mice were injected with NaHS for 18 weeks. Neonatal rat cardiomyocytes (NRCMs) were treated with high glucose, palmitate, oleate, and NaHS for 48 h. Compared to the NaHS-treated groups, in vivo and in vitro type 2 diabetic models both showed reduced intracellular H2S content, reduced cystathionine γ-lyase (CSE) expression, impaired cardiac function, decreased SERCA2a expression and decreased SERCA2a activity, reduced SUMOylation of SERCA2a, increased sentrin-specific protease 1 (SENP1) expression, and disruption of calcium homeostasis leading to activation of the mitochondrial apoptosis pathway. Compared to the NaHS-treated type 2 diabetes cellular model, overexpression of SENP1 C683A reduced the S-sulfhydration of SENP1, reduced the SUMOylation of SERCA2a, reduced the increased expression and activity of SERCA2a, and induced mitochondrial apoptosis in cardiomyocytes. These results suggested that exogenous H2S elevates SENP1 S-sulfhydration to increase SERCA2a SUMOylation, improve myocardial systolic-diastolic function, and decrease cardiomyocyte apoptosis in DCM.
Collapse
Affiliation(s)
- Shuo Peng
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Mengyi Wang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Shiwu Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Ning Liu
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Qianzhu Li
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Jiaxin Kang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Lingxue Chen
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Mingyu Li
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Kemiao Pang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Jiayi Huang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China.
| | - Dechao Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
10
|
Shaharyar MA, Bhowmik R, Al-Abbasi FA, AlGhamdi SA, Alghamdi AM, Sarkar A, Kazmi I, Karmakar S. Vaccine Formulation Strategies and Challenges Involved in RNA Delivery for Modulating Biomarkers of Cardiovascular Diseases: A Race from Laboratory to Market. Vaccines (Basel) 2023; 11:vaccines11020241. [PMID: 36851119 PMCID: PMC9963957 DOI: 10.3390/vaccines11020241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
It has been demonstrated that noncoding RNAs have significant physiological and pathological roles. Modulation of noncoding RNAs may offer therapeutic approaches as per recent findings. Small RNAs, mostly long noncoding RNAs, siRNA, and microRNAs make up noncoding RNAs. Inhibiting or promoting protein breakdown by binding to 3' untranslated regions of target mRNA, microRNAs post-transcriptionally control the pattern of gene expression. Contrarily, long non-coding RNAs perform a wider range of tasks, including serving as molecular scaffolding, decoys, and epigenetic regulators. This article provides instances of long noncoding RNAs and microRNAs that may be a biomarker of CVD (cardiovascular disease). In this paper we highlight various RNA-based vaccine formulation strategies designed to target these biomarkers-that are either currently in the research pipeline or are in the global pharmaceutical market-along with the physiological hurdles that need to be overcome.
Collapse
Affiliation(s)
- Md. Adil Shaharyar
- Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Rudranil Bhowmik
- Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Shareefa A. AlGhamdi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amira M. Alghamdi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Arnab Sarkar
- Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: (I.K.); (S.K.); Tel.: +966-543970731 (I.K.); +91-8017136385 (S.K.)
| | - Sanmoy Karmakar
- Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
- Correspondence: (I.K.); (S.K.); Tel.: +966-543970731 (I.K.); +91-8017136385 (S.K.)
| |
Collapse
|
11
|
Dong Y, Peng N, Dong L, Tan S, Zhang X. Non-coding RNAs: Important participants in cardiac fibrosis. Front Cardiovasc Med 2022; 9:937995. [PMID: 35966549 PMCID: PMC9365961 DOI: 10.3389/fcvm.2022.937995] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/24/2022] [Indexed: 11/24/2022] Open
Abstract
Cardiac remodeling is a pathophysiological process activated by diverse cardiac stress, which impairs cardiac function and leads to adverse clinical outcome. This remodeling partly attributes to cardiac fibrosis, which is a result of differentiation of cardiac fibroblasts to myofibroblasts and the production of excessive extracellular matrix within the myocardium. Non-coding RNAs mainly include microRNAs and long non-coding RNAs. These non-coding RNAs have been proved to have a profound impact on biological behaviors of various cardiac cell types and play a pivotal role in the development of cardiac fibrosis. This review aims to summarize the role of microRNAs and long non-coding RNAs in cardiac fibrosis associated with pressure overload, ischemia, diabetes mellitus, aging, atrial fibrillation and heart transplantation, meanwhile shed light on the diagnostic and therapeutic potential of non-coding RNAs for cardiac fibrosis.
Collapse
|
12
|
Updates on Cardiac Gene Therapy Research and Methods: Overview of Cardiac Gene Therapy. Methods Mol Biol 2022; 2573:3-10. [PMID: 36040582 DOI: 10.1007/978-1-0716-2707-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Gene therapy has made a significant progress in clinical translation over the past few years with several gene therapy products currently approved or anticipating approval for clinical use. Cardiac gene therapy lags behind that of other areas of diseases, with no application of cardiac gene therapy yet approved for clinical use. However, several clinical trials for gene therapy targeting the heart are underway, and innovative research studies are being conducted to close the gap. The second edition of Cardiac Gene Therapy in Methods in Molecular Biology provides protocols for cutting-edge methodologies used in these studies. In this chapter, we discuss recent updates on cardiac gene therapy studies and provide an overview of the chapters in the book.
Collapse
|
13
|
Inhibition of PLA2G4E/cPLA2 promotes survival of random skin flaps by alleviating Lysosomal membrane permeabilization-Induced necroptosis. Autophagy 2021; 18:1841-1863. [PMID: 34872436 PMCID: PMC9450981 DOI: 10.1080/15548627.2021.2002109] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Necrosis that appears at the ischemic distal end of random-pattern skin flaps increases the pain and economic burden of patients. Necroptosis is thought to contribute to flap necrosis. Lysosomal membrane permeabilization (LMP) plays an indispensable role in the regulation of necroptosis. Nonetheless, the mechanisms by which lysosomal membranes become leaky and the relationship between necroptosis and lysosomes are still unclear in ischemic flaps. Based on Western blotting, immunofluorescence, enzyme-linked immunosorbent assay, and liquid chromatography-mass spectrometry (LC-MS) analysis results, we found that LMP was presented in the ischemic distal portion of random-pattern skin flaps, which leads to disruption of lysosomal function and macroautophagic/autophagic flux, increased necroptosis, and aggravated necrosis of the ischemic flaps. Moreover, bioinformatics analysis of the LC-MS results enabled us to focus on the role of PLA2G4E/cPLA2 (phospholipase A2, group IVE) in LMP of the ischemic flaps. In vivo inhibition of PLA2G4E with an adeno-associated virus vector attenuated LMP and necroptosis, and promoted flap survival. In addition, microRNA-seq helped us determine that Mir504-5p was differentially expressed in ischemic flaps. A string of in vitro and in vivo tests was employed to verify the inhibitory effect of Mir504-5p on PLA2G4E, LMP and necroptosis. Finally, we concluded that the inhibition of PLA2G4E by Mir504-5p reduced LMP-induced necroptosis, thereby promoting the survival of random-pattern skin flaps.
Collapse
|
14
|
Crosstalk between Heart Failure and Cognitive Impairment via hsa-miR-933/RELB/CCL21 Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:2291899. [PMID: 34595235 PMCID: PMC8478533 DOI: 10.1155/2021/2291899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/26/2021] [Accepted: 08/08/2021] [Indexed: 12/15/2022]
Abstract
Background The association between heart failure (HF) and cognitive impairment has received increasing attention from scholars and researchers in recent years. However, no systematic studies have been carried out yet focused on the crosstalk between heart failure and cognitive impairment via miRNAs. Methods GSE104150, GSE53473, GSE120584, and GSE116250 with RNA-seq data and clinical data were downloaded from the GSE database. All data were statistically analysed using R software to detect DE-miRNAs and DE-mRNAs associated with both HF and cognitive impairment. Protein-protein interaction (PPI) networks were mapped, and a logistic regression model for cognitive impairment prediction was developed. Furthermore, the TTRUST database and miRWalk were used to map miRNA-transcription factor (TF) and messenger RNA (mRNA) regulatory pathways. Finally, core TFs were enriched for analysis. Results Differentially enriched DE-miRNAs and DE-mRNAs both present in HF and cognitive impairment were determined. A logistic regression model established based on DE-miRNAs was validated to have a strong performance in cognitive impairment prediction. The core miRNA-TF-mRNA pathway was formed by mapping the PPI networks associated with the two diseases. Further GSEA was performed with V-rel reticuloendotheliosis viral oncogene homolog B (RELB) as the core TF, and the retinol metabolism and gap junction pathways were analysed. Conclusions This study was the first attempt to predict the crosstalk and examine underlying mechanisms between HF and cognitive impairment applying bioinformatics. The findings suggested a potential hsa-miR-933/RELB/CCL21 regulatory axis correlated with HF and neurological disorders (or cognitive impairment), according to PPI networks.
Collapse
|
15
|
MicroRNAs and Calcium Signaling in Heart Disease. Int J Mol Sci 2021; 22:ijms221910582. [PMID: 34638924 PMCID: PMC8508866 DOI: 10.3390/ijms221910582] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 01/02/2023] Open
Abstract
In hearts, calcium (Ca2+) signaling is a crucial regulatory mechanism of muscle contraction and electrical signals that determine heart rhythm and control cell growth. Ca2+ signals must be tightly controlled for a healthy heart, and the impairment of Ca2+ handling proteins is a key hallmark of heart disease. The discovery of microRNA (miRNAs) as a new class of gene regulators has greatly expanded our understanding of the controlling module of cardiac Ca2+ cycling. Furthermore, many studies have explored the involvement of miRNAs in heart diseases. In this review, we aim to summarize cardiac Ca2+ signaling and Ca2+-related miRNAs in pathological conditions, including cardiac hypertrophy, heart failure, myocardial infarction, and atrial fibrillation. We also discuss the therapeutic potential of Ca2+-related miRNAs as a new target for the treatment of heart diseases.
Collapse
|
16
|
Marracino L, Fortini F, Bouhamida E, Camponogara F, Severi P, Mazzoni E, Patergnani S, D’Aniello E, Campana R, Pinton P, Martini F, Tognon M, Campo G, Ferrari R, Vieceli Dalla Sega F, Rizzo P. Adding a "Notch" to Cardiovascular Disease Therapeutics: A MicroRNA-Based Approach. Front Cell Dev Biol 2021; 9:695114. [PMID: 34527667 PMCID: PMC8435685 DOI: 10.3389/fcell.2021.695114] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
Dysregulation of the Notch pathway is implicated in the pathophysiology of cardiovascular diseases (CVDs), but, as of today, therapies based on the re-establishing the physiological levels of Notch in the heart and vessels are not available. A possible reason is the context-dependent role of Notch in the cardiovascular system, which would require a finely tuned, cell-specific approach. MicroRNAs (miRNAs) are short functional endogenous, non-coding RNA sequences able to regulate gene expression at post-transcriptional levels influencing most, if not all, biological processes. Dysregulation of miRNAs expression is implicated in the molecular mechanisms underlying many CVDs. Notch is regulated and regulates a large number of miRNAs expressed in the cardiovascular system and, thus, targeting these miRNAs could represent an avenue to be explored to target Notch for CVDs. In this Review, we provide an overview of both established and potential, based on evidence in other pathologies, crosstalks between miRNAs and Notch in cellular processes underlying atherosclerosis, myocardial ischemia, heart failure, calcification of aortic valve, and arrhythmias. We also discuss the potential advantages, as well as the challenges, of using miRNAs for a Notch-based approach for the diagnosis and treatment of the most common CVDs.
Collapse
Affiliation(s)
- Luisa Marracino
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | | | - Esmaa Bouhamida
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesca Camponogara
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Paolo Severi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Elisa Mazzoni
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Simone Patergnani
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Emanuele D’Aniello
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Roberta Campana
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
| | | | - Paola Rizzo
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
| |
Collapse
|
17
|
Mahjoubin-Tehran M, Atkin SL, Bezsonov EE, Jamialahmadi T, Sahebkar A. Harnessing the Therapeutic Potential of Decoys in Non-Atherosclerotic Cardiovascular Diseases: State of the Art. J Cardiovasc Dev Dis 2021; 8:103. [PMID: 34564121 PMCID: PMC8467637 DOI: 10.3390/jcdd8090103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/06/2021] [Accepted: 08/20/2021] [Indexed: 01/30/2023] Open
Abstract
Cardiovascular disease (CVD) is the main cause of global death, highlighting the fact that conventional therapeutic approaches for the treatment of CVD patients are insufficient, and there is a need to develop new therapeutic approaches. In recent years, decoy technology, decoy oligodeoxynucleotides (ODN), and decoy peptides show promising results for the future treatment of CVDs. Decoy ODN inhibits transcription by binding to the transcriptional factor, while decoy peptide neutralizes receptors by binding to the ligands. This review focused on studies that have investigated the effects of decoy ODN and decoy peptides on non-atherosclerotic CVD.
Collapse
Affiliation(s)
- Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran;
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Evgeny E. Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia;
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, Moscow, Russia
- Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran;
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| |
Collapse
|
18
|
Trophoblasts Modulate the Ca 2+ Oscillation and Contraction of Myometrial Smooth Muscle Cells by Small Extracellular Vesicle- (sEV-) Mediated Exporting of miR-25-3p during Premature Labor. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8140667. [PMID: 34413928 PMCID: PMC8369173 DOI: 10.1155/2021/8140667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/06/2021] [Accepted: 07/26/2021] [Indexed: 12/31/2022]
Abstract
The placenta could transmit information to the maternal circulation via the secretion of small extracellular vesicles (sEVs), but little is known about whether and how these sEVs participate in premature labor (PTL). We speculate that miRNA plays an important role in sEV-mediated fetal-maternal information transmission. The present study was aimed at investigating whether the placenta can regulate the contraction of the maternal myometrium via sEV-mediated transmit of miR-25-3p during PTL. The expression of miR-25-3p and its targets Cav3.2 and SERCA2a in clinical samples, cells, and animal specimens was detected. The role of miR-25-3p was observed in the LPS-induced preterm labor mouse model. The sEVs from HTR-8/SVneo cells were characterized by transmission electron microscopy and nanoparticle tracking analysis. The Ca2+ oscillation in HMSMs was analyzed by an intracellular Ca2+ change tracking assay on a confocal microscope. The contraction of HMSMs was detected by a collagen matrix contraction assay. We found that miR-25-3p can directly bind to the 3′UTR of Cav3.2 and SERCA2a. The miR-25-3p level was decreased, and the expression of its targets Cav3.2 and SERCA2a was increased in the placenta and myometrium tissues of PTL patients. Forced upregulation of miR-25-3p reduced the oxidative stress and inflammation responses and the incidence of PTL in LPS-treated mice. The expression of miR-25-3p was not changed in LPS-stimulated human myometrial smooth muscle cells (HMSMs) but was strongly reduced in the trophoblast cell and its sEVs. Additionally, the trophoblast cell line HTR-8/SVneo could transmit miR-25-3p into HMSMs via sEVs. The sEVs derived from LPS-stimulated trophoblasts upregulated the expression of Cav3.2 and SERCA2a and triggered Ca2+ oscillation as well as the contraction of HMSMs; these effects were partially reversed by the overexpression of miR-25-3p in the trophoblasts. Further, the upregulation of miR-25-3p induced changes of Ca2+ oscillation and contraction of HMSMs mediated by sEVs which were also abrogated by the knockdown of miR-25-3p in HMSMs. The results demonstrated that miR-25-3p plays a crucial role in PTL via Cav3.2- and SERCA2a-mediated Ca2+ oscillation and contraction of HMSMs. PTL seems to be related to the decreased exosomal miR-25-3p content transmitted by the trophoblasts under inflammatory conditions.
Collapse
|
19
|
Mahjoubin-Tehran M, Rezaei S, Atkin SL, Montecucco F, Sahebkar A. Decoys as potential therapeutic tools for diabetes. Drug Discov Today 2021; 26:1669-1679. [PMID: 33862194 DOI: 10.1016/j.drudis.2021.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 01/15/2021] [Accepted: 04/06/2021] [Indexed: 10/21/2022]
Abstract
Current therapeutic approaches for diabetes are focused on improving glycemic control to prevent diabetes-related complications, but such approached are not completely successful. Decoy technologies such as decoy oligodeoxynucleotides (ODNs) and decoy peptides have emerged as therapeutic tools in diabetes. Decoy ODNs carry a DNA recognition motif for the binding of transcription factors in order to trap them and block their effects, whereas decoy peptides mimic the binding structure of the receptor protein, bind to the docking site of the target ligand, and prevent the interaction of the ligand and receptor. This review summarizes the technologies that have been developed to date and the studies that have investigated the therapeutic effects of decoy ODNs and peptides in diabetes.
Collapse
Affiliation(s)
- Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Rezaei
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Non-Coding RNAs in the Cardiac Action Potential and Their Impact on Arrhythmogenic Cardiac Diseases. HEARTS 2021. [DOI: 10.3390/hearts2030026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cardiac arrhythmias are prevalent among humans across all age ranges, affecting millions of people worldwide. While cardiac arrhythmias vary widely in their clinical presentation, they possess shared complex electrophysiologic properties at cellular level that have not been fully studied. Over the last decade, our current understanding of the functional roles of non-coding RNAs have progressively increased. microRNAs represent the most studied type of small ncRNAs and it has been demonstrated that miRNAs play essential roles in multiple biological contexts, including normal development and diseases. In this review, we provide a comprehensive analysis of the functional contribution of non-coding RNAs, primarily microRNAs, to the normal configuration of the cardiac action potential, as well as their association to distinct types of arrhythmogenic cardiac diseases.
Collapse
|
21
|
Xu M, Zhang K, Song J. Targeted Therapy in Cardiovascular Disease: A Precision Therapy Era. Front Pharmacol 2021; 12:623674. [PMID: 33935716 PMCID: PMC8085499 DOI: 10.3389/fphar.2021.623674] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Targeted therapy refers to exploiting the specific therapeutic drugs against the pathogenic molecules (a protein or a gene) or cells. The drug specifically binds to disease-causing molecules or cells without affecting normal tissue, thus enabling personalized and precision treatment. Initially, therapeutic drugs included antibodies and small molecules, (e.g. nucleic acid drugs). With the advancement of the biology technology and immunotherapy, the gene editing and cell editing techniques are utilized for the disease treatment. Currently, targeted therapies applied to treat cardiovascular diseases (CVDs) mainly include protein drugs, gene editing technologies, nucleic acid drugs and cell therapy. Although targeted therapy has demonstrated excellent efficacy in pre-clinical and clinical trials, several limitations need to be recognized and overcome in clinical application, (e.g. off-target events, gene mutations, etc.). This review introduces the mechanisms of different targeted therapies, and mainly describes the targeted therapy applied in the CVDs. Furthermore, we made comparative analysis to clarify the advantages and disadvantages of different targeted therapies. This overview is expected to provide a new concept to the treatment of the CVDs.
Collapse
Affiliation(s)
- Mengda Xu
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kailun Zhang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Kailun Zhang, ; Jiangping Song,
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,*Correspondence: Kailun Zhang, ; Jiangping Song,
| |
Collapse
|
22
|
Zeng N, Wen YH, Pan R, Yang J, Yan YM, Zhao AZ, Zhu JN, Fang XH, Shan ZX. Dickkopf 3: a Novel Target Gene of miR-25-3p in Promoting Fibrosis-Related Gene Expression in Myocardial Fibrosis. J Cardiovasc Transl Res 2021; 14:1051-1062. [PMID: 33723747 DOI: 10.1007/s12265-021-10116-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/25/2021] [Indexed: 10/21/2022]
Abstract
Increasing evidence has shown that microRNAs (miRNAs) participate in cardiac fibrosis. We aimed to elucidate the effect of miRNA miR-25-3p on cardiac fibrosis. MiRNA microarray was used to profile miRNAs in the myocardium of angiotensin-II (Ang-II)-infused mice. Effect of miR-25-3p on expression of fibrosis-related genes, including Col1a1, Col3a1, and Acta2, was investigated both in vitro and in vivo. MiR-25-3p was shown increased in the myocardium of Ang-II-infused mice and patients with heart failure. MiR-25-3p enhanced fibrosis-related gene expression in mouse cardiac fibroblasts (mCFs) and in the myocardium of Ang-II-infused mice. Dickkopf 3 (Dkk3) was identified as a target gene of miR-25-3p, and Dkk3 could ameliorate Smad3 activation and fibrosis-related gene expression via enhancing Smad7 expression in mCFs. Additionally, NF-κB signal was proven to mediate upregulation of miR-25-3p in cardiac fibrosis. Our findings suggest that miR-25-3p enhances cardiac fibrosis by suppressing Dkk3 to activate Smad3 and fibrosis-related gene expression.
Collapse
Affiliation(s)
- Ni Zeng
- School of Medicine, South China University of Technology, Guangzhou, 510632, China
| | - Yi-Hong Wen
- School of Medicine, South China University of Technology, Guangzhou, 510632, China
| | - Rong Pan
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, 510632, China
| | - Jing Yang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yu-Min Yan
- School of Pharmacy, Southern Medical University, Guangzhou, 510515, China
| | - An-Zhi Zhao
- School of Pharmacy, Southern Medical University, Guangzhou, 510515, China
| | - Jie-Ning Zhu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou, 510080, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Xian-Hong Fang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou, 510080, China.
| | - Zhi-Xin Shan
- School of Medicine, South China University of Technology, Guangzhou, 510632, China. .,School of Pharmacy, Southern Medical University, Guangzhou, 510515, China. .,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| |
Collapse
|
23
|
Knockdown of Long Noncoding RNA SNHG14 Protects H9c2 Cells Against Hypoxia-induced Injury by Modulating miR-25-3p/KLF4 Axis in Vitro. J Cardiovasc Pharmacol 2021; 77:334-342. [PMID: 33278191 DOI: 10.1097/fjc.0000000000000965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/05/2020] [Indexed: 11/25/2022]
Abstract
ABSTRACT Cyanotic congenital heart disease (CCHD) is the main cause of death in infants worldwide. Long noncoding RNAs (lncRNAs) have been pointed to exert crucial roles in development of CHD. The current research is designed to illuminate the impact and potential mechanism of lncRNA SNHG14 in CCHD in vitro. The embryonic rat ventricular myocardial cells (H9c2 cells) were exposed to hypoxia to establish the model of CCHD in vitro. Quantitative real-time polymerase chain reaction was conducted to examine relative expressions of SNHG14, miR-25-3p, and KLF4. Cell viability was determined by the MTT assay. Lactate dehydrogenase (LDH) was measured by an LDH assay kit. Apoptosis-related proteins (Bax and Bcl-2) and KLF4 were detected by Western Blot. The targets of SNHG14 and miR-25-3p were verified by the dual-luciferase reporter assay. SNHG14 and KLF4 were upregulated, whereas miR-25-3p was downregulated in hypoxia-induced H9c2 cells and cardiac tissues of patients with CCHD compared with their controls. Knockdown of SNHG14 or overexpression of miR-25-3p facilitated cell viability, while depressing cell apoptosis and release of LDH in hypoxia-induced H9c2 cells. MiR-25-3p was a target of SNHG14 and inversely modulated by SNHG14. MiR-25-3p could directly target KLF4 and negatively regulate expression of KLF4. Repression of miR-25-3p or overexpression of KLF4 reversed the suppression impacts of sh-SNHG14 on cell apoptosis and release of LDH as well as the promotion impact of sh-SNHG14 on cell viability in hypoxia-induced H9c2 cells. Sh-SNHG14 protected H9c2 cells against hypoxia-induced injury by modulating miR-25-3p/KLF4 axis in vitro.
Collapse
|
24
|
MicroRNAs Regulating Autophagy in Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1208:191-264. [PMID: 34260028 DOI: 10.1007/978-981-16-2830-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Social and economic impacts of neurodegenerative diseases (NDs) become more prominent in our constantly aging population. Currently, due to the lack of knowledge about the aetiology of most NDs, only symptomatic treatment is available for patients. Hence, researchers and clinicians are in need of solid studies on pathological mechanisms of NDs. Autophagy promotes degradation of pathogenic proteins in NDs, while microRNAs post-transcriptionally regulate multiple signalling networks including autophagy. This chapter will critically discuss current research advancements in the area of microRNAs regulating autophagy in NDs. Moreover, we will introduce basic strategies and techniques used in microRNA research. Delineation of the mechanisms contributing to NDs will result in development of better approaches for their early diagnosis and effective treatment.
Collapse
|
25
|
The Protective Effect of Qishen Granule on Heart Failure after Myocardial Infarction through Regulation of Calcium Homeostasis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:1868974. [PMID: 33149749 PMCID: PMC7603572 DOI: 10.1155/2020/1868974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/26/2020] [Accepted: 09/22/2020] [Indexed: 12/16/2022]
Abstract
Qishen granule (QSG) is a frequently prescribed traditional Chinese medicine formula, which improves heart function in patients with heart failure (HF). However, the cardioprotective mechanisms of QSG have not been fully understood. The current study aimed to elucidate whether the effect of QSG is mediated by ameliorating cytoplasmic calcium (Ca2+) overload in cardiomyocytes. The HF rat model was induced by left anterior descending (LAD) artery ligation surgery. Rats were randomly divided into sham, model, QSG-low dosage (QSG-L) treatment, QSG-high dosage (QSG-H) treatment, and positive drug (diltiazem) treatment groups. 28 days after surgery, cardiac functions were assessed by echocardiography. Levels of norepinephrine (NE) and angiotensin II (AngII) in the plasma were evaluated. Expressions of critical proteins in the calcium signaling pathway, including cell membrane calcium channel CaV1.2, sarcoendoplasmic reticulum ATPase 2a (SERCA2a), calcium/calmodulin-dependent protein kinase type II (CaMKII), and protein phosphatase calcineurin (CaN), were measured by Western blotting (WB) and immunohistochemistry (IHC). Echocardiography showed that left ventricular ejection fraction (EF) and fractional shortening (FS) value significantly decreased in the model group compared to the sham group, and illustrating heart function was severely impaired. Furthermore, levels of NE and AngII in the plasma were dramatically increased. Expressions of CaV1.2, CaMKII, and CaN in the cardiomyocytes were upregulated, and expressions of SERCA2a were downregulated in the model group. After treatment with QSG, both EF and FS values were increased. QSG significantly reduced levels of NE and AngII in the plasma. In particular, QSG prevented cytoplasmic Ca2+ overload by downregulating expression of CaV1.2 and upregulating expression of SERCA2a. Meanwhile, expressions of CaMKII and CaN were inhibited by QSG treatment. In conclusion, QSG could effectively promote heart function in HF rats by restoring cardiac Ca2+ homeostasis. These findings revealed novel therapeutic mechanisms of QSG and provided potential targets in the treatment of HF.
Collapse
|
26
|
Zhang Z, Liu Q, Yang J, Yao H, Fan R, Cao C, Liu C, Zhang S, Lei X, Xu S. The proteomic profiling of multiple tissue damage in chickens for a selenium deficiency biomarker discovery. Food Funct 2020; 11:1312-1321. [PMID: 32022057 DOI: 10.1039/c9fo02861g] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past decades, substantial advances have been made in both the early diagnosis and accurate prognosis of numerous cancers because of the impressive development of novel proteomic strategies. Selenium (Se) is an essential trace element in humans and animals. Se deficiency could lead to Keshan disease in humans, mulberry heart disease in pigs and damage of tissues including cardiac injury, apoptosis in the liver, reduction in the immune responses in spleen and cerebral lesions in chickens. However, it is well know that plasma biomarkers are not specific and also show alterations in various diseases including those caused by Se deficiency. Therefore, new definition biomarkers are needed to improve disease surveillance and reduce unnecessary chicken losses due to Se deficiency. To identify new biomarkers for Se deficiency, we performed exploratory heart, liver, spleen, muscle, vein, and artery proteomic screens to further validate the biomarkers using Venn analysis, GO enrichment, heatmap analysis, and IPA analysis. Based on the bioinformatics methods mentioned above, we found that differentially expressed genes and proteins are enriched to the PI3K/AKT/mTOR signal pathway and insulin pathway. We further used western blot to detect the expression of proteins related to the two pathways. Results showed that the components of the PI3K/AKT/mTOR signal pathway were definitely decreased in heart, liver, spleen, muscle, vein and artery tissues in the Se deficient group. Expression IGF and IGFBP2 of the insulin pathway were differentially increased in the heart, liver, and spleen in Se deficient group samples and decreased in muscle and artery. In conclusion, 5 proteins, namely PI3K, AKT, mTOR, IGF, and IGFBP2, were differentially expressed, which could be potentially useful Se deficient biomarkers. In the present study, proteomic profiling was used to elucidate protein biomarkers that distinguished Se deficient samples from the controls, which might provide a new direction for the diagnosis and targeted treatment induced by Se deficiency in chickens.
Collapse
Affiliation(s)
- Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China. and Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Qi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| | - Jie Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| | - Haidong Yao
- Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | - Ruifeng Fan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, P. R. China
| | - Changyu Cao
- College of Life and Science, Foshan University, Foshan, 528000, P. R. China
| | - Ci Liu
- College of Animal Technology, Shanxi Agricultural University, Jinzhong, 030600, P. R. China
| | - Sheng Zhang
- Institute of Biotechnology, Cornell University, Ithaca, NY 14583, USA
| | - Xingen Lei
- Department of Animal Science, Cornell University, Ithaca, NY 14583, USA
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China. and Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| |
Collapse
|
27
|
A tangled tale of microRNA and cardiac fibrosis. Clin Sci (Lond) 2020; 133:2217-2220. [PMID: 31722012 DOI: 10.1042/cs20190866] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022]
Abstract
Cardiac fibrosis is important for wound healing, regeneration and producing the extracellular matrix (ECM) that provides the scaffold for cells. In pathological situations, fibroblasts are activated and remodel the ECM. In volume 133, issue 17 of Clinical Science, Yang et al. discovered that the miR-214-3p/NLRC5 axis is important for fibroblast-to-myofibroblast transition (FMT) and ECM remodelling in a pressure overload model of fibrosis [Clin. Sci. (2019) 133(17), 1845-1856]. This discovery helps to explain the complicated regulation of cardiac fibrosis. It also underscores the need for more investigation into the mechanisms of cardiac fibrosis to develop better diagnostic modalities and therapeutic options in heart failure.
Collapse
|
28
|
Li Z, Li H, Liu B, Luo J, Qin X, Gong M, Shi B, Wei Y. Inhibition of miR-25 attenuates doxorubicin-induced apoptosis, reactive oxygen species production and DNA damage by targeting PTEN. Int J Med Sci 2020; 17:1415-1427. [PMID: 32624698 PMCID: PMC7330660 DOI: 10.7150/ijms.41980] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Doxorubicin (DOX) is one of the widely used anti-cancer drugs, whereas it can induce irreversible cardiac injury in a dose-dependent manner which limits its utility in clinic. Our study aimed to investigate the relationship between miR-25 and DOX-induced cardiac injury and its underlying mechanism. Methods: Mice and H9c2 cells were exposed to DOX. The overexpressed or knockdown of miR-25 in H9c2 cells was achieved by miR-25 mimic or inhibitor and the efficiency of transfection was identified by qRT-PCR or Western blotting. Cell viability, apoptotic cell rate, and levels of apoptosis-related proteins were determined by CCK-8, flow cytometry, and Western blotting, respectively. Furthermore, Western blotting and immunofluorescence staining (IF) were performed to assess the expression levels of reactive oxygen species and degree of DNA damage. Results: As a result, DOX significantly upregulated miR-25 expression in mice and H9c2 cells and reduced cell viability and increased cell apoptosis in vitro and in vivo. miR-25 overexpression expedited cell injury induced by DOX in H9c2 cells demonstrated by the increased cell apoptosis and reactive oxygen species (ROS) production, whereas miR-25 inhibition attenuated the cell injury. Furthermore, miR-25 negatively controlled the expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN). Intervention the expression of PTEN using si-PTEN reversed the beneficial effects of miR-25 inhibition on DOX-injured H9c2 cells. Conclusion: In conclusion, this study demonstrated that miR-25 is involved in DOX-induced cell damage through the regulation of PTEN expression.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Hongqiang Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Baoxin Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Jiachen Luo
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Xiaoming Qin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Mengmeng Gong
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Beibei Shi
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Yidong Wei
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| |
Collapse
|
29
|
Ooi JYY, Bernardo BC. Translational Potential of Non-coding RNAs for Cardiovascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:343-354. [PMID: 32285423 DOI: 10.1007/978-981-15-1671-9_21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jenny Y Y Ooi
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Bianca C Bernardo
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
30
|
Xu M, Li J, Xie J, He R, Su Q, Gao G, Tai PW. High-Throughput Quantification of In Vivo Adeno-Associated Virus Transduction with Barcoded Non-Coding RNAs. Hum Gene Ther 2019; 30:946-956. [PMID: 31072208 PMCID: PMC6703241 DOI: 10.1089/hum.2018.253] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 04/12/2019] [Indexed: 02/06/2023] Open
Abstract
Recombinant adeno-associated viruses (rAAVs) have become favorable gene delivery vehicles for expressing therapeutic transgenes. Capsid engineering efforts to produce novel AAVs with improved transduction efficiencies, unique tissue specificities, and reduced host immunities are a direct response to the high demand for treatment needs that preexisting rAAVs cannot currently fulfill. New AAV capsids discovered by directed evolution methods, in silico design, or from natural proviral sequences ultimately require extensive characterization in relevant in vivo models. Consequently, quantitative screening of candidate capsid libraries now requires reliable high-throughput sequencing approaches. In this study, we have developed a vector/transgene tracking system that employs the indexing of a non-coding RNA. Specifically, a barcoded Tough Decoy (bcTuD) that express highly stable RNA transcripts that can be used as readouts for transduction efficiency. The pseudo-hairpin structure of the bcTuD contains a variable region that is amenable to barcode insertion, which can be detected by target amplicon sequencing. The described approach, named AAV-bcTuD screening, offers a new alternative for in vivo assessment of rAAV that can accurately quantify vector genomes and transcript abundances in tissues, as exampled by the demonstration in liver and brain infections. Proof-of-concept is provided to show that vector genome and transcript detection in tissues with this method is accurate and consistent for a vector dose range of upwards to four logs in a mixed vector injection, showing that this technique is robust, sensitive, and applicable for multiplexed screening of capsid performance in vivo.
Collapse
Affiliation(s)
- Meiyu Xu
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jia Li
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jun Xie
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Ran He
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Qin Su
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Phillip W.L. Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
31
|
Antunes JC, Benarroch L, Moraes FC, Juenet M, Gross MS, Aubart M, Boileau C, Caligiuri G, Nicoletti A, Ollivier V, Chaubet F, Letourneur D, Chauvierre C. Core-Shell Polymer-Based Nanoparticles Deliver miR-155-5p to Endothelial Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:210-222. [PMID: 31265949 PMCID: PMC6610682 DOI: 10.1016/j.omtn.2019.05.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022]
Abstract
Heart failure occurs in over 30% of the worldwide population and most commonly originates from cardiovascular diseases such as myocardial infarction. microRNAs (miRNAs) target and silence specific mRNAs, thereby regulating gene expression. Because the endogenous miR-155-5p has been ascribed to vasculoprotection, loading it onto positively charged, core-shell poly(isobutylcyanoacrylate) (PIBCA)-polysaccharide nanoparticles (NPs) was attempted. NPs showed a decrease (p < 0.0001) in surface electrical charge (ζ potential), with negligible changes in size or shape when loaded with the anionic miR-155-5p. Presence of miR-155-5p in loaded NPs was further quantified. Cytocompatibility up to 100 μg/mL of NPs for 2 days with human coronary artery endothelial cells (hCAECs) was documented. NPs were able to enter hCAECs and were localized in the endoplasmic reticulum (ER). Expression of miR-155-5p was increased within the cells by 75-fold after 4 hours of incubation (p < 0.05) and was still noticeable at day 2. Differences between loaded NP-cultured cells and free miRNA, at days 1 (p < 0.05) and 2 (p < 0.001) suggest the ability of prolonged load release in physiological conditions. Expression of miR-155-5p downstream target BACH1 was decreased in the cells by 4-fold after 1 day of incubation (p < 0.05). This study is a first proof of concept that miR-155-5p can be loaded onto NPs and remain intact and biologically active in endothelial cells (ECs). These nanosystems could potentially increase an endogenous cytoprotective response and decrease damage within infarcted hearts.
Collapse
Affiliation(s)
- Joana C Antunes
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Louise Benarroch
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Fernanda C Moraes
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Maya Juenet
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Marie-Sylvie Gross
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Mélodie Aubart
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Catherine Boileau
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Giuseppina Caligiuri
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Antonino Nicoletti
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Véronique Ollivier
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Frédéric Chaubet
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Didier Letourneur
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Cédric Chauvierre
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France.
| |
Collapse
|
32
|
Katz MG, Fargnoli AS, Hajjar RJ, Jeong D. MicroRNA-25 upregulation protects spinal cords, yet is bad for the heart: The dark side of noncoding RNAS. J Thorac Cardiovasc Surg 2019; 158:e87-e88. [PMID: 31003740 DOI: 10.1016/j.jtcvs.2019.03.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 11/26/2022]
Affiliation(s)
- Michael G Katz
- Department of Cardiology, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anthony S Fargnoli
- Department of Cardiology, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Roger J Hajjar
- Department of Cardiology, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Dongtak Jeong
- Department of Cardiology, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
33
|
Oh JG, Jang SP, Yoo J, Lee MA, Lee SH, Lim T, Jeong E, Kho C, Kook H, Hajjar RJ, Park WJ, Jeong D. Role of the PRC2-Six1-miR-25 signaling axis in heart failure. J Mol Cell Cardiol 2019; 129:58-68. [DOI: 10.1016/j.yjmcc.2019.01.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/25/2018] [Accepted: 01/21/2019] [Indexed: 01/14/2023]
|
34
|
Wadley GD, Lamon S, Alexander SE, McMullen JR, Bernardo BC. Noncoding RNAs regulating cardiac muscle mass. J Appl Physiol (1985) 2018; 127:633-644. [PMID: 30571279 DOI: 10.1152/japplphysiol.00904.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Noncoding RNAs, including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs) play roles in the development and homeostasis of nearly every tissue of the body, including the regulation of processes underlying heart growth. Cardiac hypertrophy can be classified as either physiological (beneficial heart growth) or pathological (detrimental heart growth), the latter of which results in impaired cardiac function and heart failure and is predictive of a higher incidence of death due to cardiovascular disease. Several miRNAs have a functional role in exercise-induced cardiac hypertrophy, while both miRNAs and lncRNAs are heavily involved in pathological heart growth and heart failure. The latter have the potential to act as an endogenous sponge RNA and interact with specific miRNAs to control cardiac hypertrophy, adding another level of complexity to our understanding of the regulation of cardiac muscle mass. In addition to tissue-specific effects, ncRNA-mediated tissue cross talk occurs via exosomes. In particular, miRNAs can be internalized in exosomes and secreted from various cardiac and vascular cell types to promote angiogenesis, as well as protection and repair of ischemic tissues. ncRNAs hold promising therapeutic potential to protect the heart against ischemic injury and aid in regeneration. Numerous preclinical studies have demonstrated the therapeutic potential of ncRNAs, specifically miRNAs, for the treatment of cardiovascular disease. Most of these studies employ antisense oligonucleotides to inhibit miRNAs of interest; however, off-target effects often limit their potential to be translated to the clinic. In this context, approaches using viral and nonviral delivery tools are promising means to provide targeted delivery in vivo.
Collapse
Affiliation(s)
- Glenn D Wadley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Séverine Lamon
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Sarah E Alexander
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia.,Department of Medicine, Monash University, Clayton, Victoria, Australia.,Department of Physiology, Monash University, Clayton, Victoria, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - Bianca C Bernardo
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Victoria, Australia
| |
Collapse
|
35
|
Hooykaas MJG, Soppe JA, De Buhr HM, Kruse E, Wiertz EJHJ, Lebbink RJ. RNA accessibility impacts potency of Tough Decoy microRNA inhibitors. RNA Biol 2018; 15:1410-1419. [PMID: 30339041 PMCID: PMC6284568 DOI: 10.1080/15476286.2018.1537746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
MicroRNAs (miRNAs) are small RNA molecules that post-transcriptionally regulate gene expression through silencing of complementary target mRNAs. miRNAs are involved in many biological processes, including cell proliferation, differentiation, cell signaling and cellular defense responses to infection. Strategies that allow for strong and stable suppression of specific microRNA activity are needed to study miRNA functions and to develop therapeutic intervention strategies aimed at interfering with miRNA activity in vivo. One of these classes of miRNA inhibitors are Tough Decoys (TuD) RNAs, which comprise of an imperfect RNA hairpin structure that harbors two opposing miRNA binding sites. Upon developing TuDs targeting Epstein-Barr virus miRNAs, we observed a strong variation in inhibitory potential between different TuD RNAs targeting the same miRNA. We show that the composition of the 'bulge' sequence in the miRNA binding sites has a strong impact on the inhibitory potency of the TuD. Our data implies that miRNA inhibition correlates with the thermodynamic properties of the TuD and that design aimed at lowering the TuD opening energy increases TuD potency. Our study provides specific guidelines for the design and construction of potent decoy-based miRNA inhibitors, which may be used for future therapeutic intervention strategies.
Collapse
Affiliation(s)
- Marjolein J G Hooykaas
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Jasper A Soppe
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Hendrik M De Buhr
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Elisabeth Kruse
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Emmanuel J H J Wiertz
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Robert J Lebbink
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| |
Collapse
|
36
|
Bernardo BC, Gregorevic P, Ritchie RH, McMullen JR. Generation of MicroRNA-34 Sponges and Tough Decoys for the Heart: Developments and Challenges. Front Pharmacol 2018; 9:1090. [PMID: 30298011 PMCID: PMC6160554 DOI: 10.3389/fphar.2018.01090] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Heart failure (HF) is a debilitating and deadly chronic disease, with almost 50% of patients with HF dying within 5 years of diagnosis. With limited effective therapies to treat or cure HF, new therapies are greatly needed. microRNAs (miRNAs) are small non-coding RNA molecules that are powerful regulators of gene expression and play a key role in almost every biological process. Disruptions in miRNA gene expression has been functionally linked to numerous diseases, including cardiovascular disease. Molecular tools for manipulating miRNA activity have been developed, and there is evidence from preclinical studies demonstrating the potential of miRNAs to be therapeutic targets for cardiovascular disease. For clinical application, miRNA sponges and tough decoys have been developed for more stable suppression and targeted delivery of the miRNA of choice. The aim of this study was to generate miRNA sponges and tough decoys to target miR-34 in the mouse heart. We present data to show that using both approaches we were unable to get significant knockdown of miR-34 or regulate miR-34 target genes in the heart in vivo. We also review recent applications of this method in the heart and discuss further considerations for optimisation in construct design and testing, and the obstacles to be overcome before they enter the clinic.
Collapse
Affiliation(s)
- Bianca C Bernardo
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Paul Gregorevic
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Rebecca H Ritchie
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia.,Department of Medicine, Monash University, Clayton, VIC, Australia.,Department of Physiology, Monash University, Clayton, VIC, Australia.,Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|