1
|
Udugampolage NS, Frolova S, Taurino J, Pini A, Martelli F, Voellenkle C. Coding and Non-Coding Transcriptomic Landscape of Aortic Complications in Marfan Syndrome. Int J Mol Sci 2024; 25:7367. [PMID: 39000474 PMCID: PMC11242319 DOI: 10.3390/ijms25137367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Marfan syndrome (MFS) is a rare congenital disorder of the connective tissue, leading to thoracic aortic aneurysms (TAA) and dissection, among other complications. Currently, the most efficient strategy to prevent life-threatening dissection is preventive surgery. Periodic imaging applying complex techniques is required to monitor TAA progression and to guide the timing of surgical intervention. Thus, there is an acute demand for non-invasive biomarkers for diagnosis and prognosis, as well as for innovative therapeutic targets of MFS. Unraveling the intricate pathomolecular mechanisms underlying the syndrome is vital to address these needs. High-throughput platforms are particularly well-suited for this purpose, as they enable the integration of different datasets, such as transcriptomic and epigenetic profiles. In this narrative review, we summarize relevant studies investigating changes in both the coding and non-coding transcriptome and epigenome in MFS-induced TAA. The collective findings highlight the implicated pathways, such as TGF-β signaling, extracellular matrix structure, inflammation, and mitochondrial dysfunction. Potential candidates as biomarkers, such as miR-200c, as well as therapeutic targets emerged, like Tfam, associated with mitochondrial respiration, or miR-632, stimulating endothelial-to-mesenchymal transition. While these discoveries are promising, rigorous and extensive validation in large patient cohorts is indispensable to confirm their clinical relevance and therapeutic potential.
Collapse
Affiliation(s)
| | - Svetlana Frolova
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
- Department of Biosciences, University of Milan, 20122 Milan, Italy
| | - Jacopo Taurino
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, 20097 Milan, Italy; (N.S.U.); (J.T.); (A.P.)
| | - Alessandro Pini
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, 20097 Milan, Italy; (N.S.U.); (J.T.); (A.P.)
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
| |
Collapse
|
2
|
Greco S, Made' A, Mutoli M, Zhang L, Piella SN, Vausort M, Lumley AI, Beltrami AP, Srivastava PK, Milani V, Boveri S, Ranucci M, Renna LV, Firat H, Bruno A, Spinetti G, Emanueli C, Devaux Y, Martelli F. HCG18, LEF1AS1 and lncCEACAM21 as biomarkers of disease severity in the peripheral blood mononuclear cells of COVID-19 patients. J Transl Med 2023; 21:758. [PMID: 37884975 PMCID: PMC10605335 DOI: 10.1186/s12967-023-04497-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/01/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Even after 3 years from SARS-CoV-2 identification, COVID-19 is still a persistent and dangerous global infectious disease. Significant improvements in our understanding of the disease pathophysiology have now been achieved. Nonetheless, reliable and accurate biomarkers for the early stratification of COVID-19 severity are still lacking. Long noncoding RNAs (LncRNAs) are ncRNAs longer than 200 nucleotides, regulating the transcription and translation of protein-coding genes and they can be found in the peripheral blood, thus holding a promising biomarker potential. Specifically, peripheral blood mononuclear cells (PBMCs) have emerged as a source of indirect biomarkers mirroring the conditions of tissues: they include monocytes, B and T lymphocytes, and natural killer T cells (NKT), being highly informative for immune-related events. METHODS We profiled by RNA-Sequencing a panel of 2906 lncRNAs to investigate their modulation in PBMCs of a pilot group of COVID-19 patients, followed by qPCR validation in 111 hospitalized COVID-19 patients. RESULTS The levels of four lncRNAs were found to be decreased in association with COVID-19 mortality and disease severity: HLA Complex Group 18-242 and -244 (HCG18-242 and HCG18-244), Lymphoid Enhancer Binding Factor 1-antisense 1 (LEF1-AS1) and lncCEACAM21 (i.e. ENST00000601116.5, a lncRNA in the CEACAM21 locus). Interestingly, these deregulations were confirmed in an independent patient group of hospitalized patients and by the re-analysis of publicly available single-cell transcriptome datasets. The identified lncRNAs were expressed in all of the PBMC cell types and inversely correlated with the neutrophil/lymphocyte ratio (NLR), an inflammatory marker. In vitro, the expression of LEF1-AS1 and lncCEACAM21 was decreased upon THP-1 monocytes exposure to a relevant stimulus, hypoxia. CONCLUSION The identified COVID-19-lncRNAs are proposed as potential innovative biomarkers of COVID-19 severity and mortality.
Collapse
Affiliation(s)
- Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy.
| | - Alisia Made'
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy
| | - Martina Mutoli
- IRCCS MultiMedica, Via Fantoli 16/15, 20138, Milan, Italy
| | - Lu Zhang
- Bioinformatics Platform, Data Integration and Analysis Unit, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
| | - Santiago Nicolas Piella
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy
| | - Mélanie Vausort
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
| | - Andrew I Lumley
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
| | | | - Prashant Kumar Srivastava
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, London, W12 0NN, England, UK
| | - Valentina Milani
- Laboratory of Biostatistics and Data Management, Scientific Directorate, IRCCS Policlinico San Donato, 20097, San Donato Milanese, Milan, Italy
| | - Sara Boveri
- Laboratory of Biostatistics and Data Management, Scientific Directorate, IRCCS Policlinico San Donato, 20097, San Donato Milanese, Milan, Italy
| | - Marco Ranucci
- Department of Cardiovascular Anesthesia and ICU, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy
| | - Laura Valentina Renna
- Biobank BioCor, IRCCS-Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy
| | | | - Antonino Bruno
- IRCCS MultiMedica, Via Fantoli 16/15, 20138, Milan, Italy
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Via Monte Generoso 71, 21100, Varese, Italy
| | - Gaia Spinetti
- IRCCS MultiMedica, Via Fantoli 16/15, 20138, Milan, Italy
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, London, W12 0NN, England, UK.
- National Heart & Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, SW3 6LY, UK.
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445, Strassen, Luxembourg.
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Milan, Italy.
| |
Collapse
|
3
|
Li JC, Jia J, Dong L, Hu ZJ, Huang XR, Wang HL, Wang L, Yang SJ, Lan HY. Angiotensin II mediates hypertensive cardiac fibrosis via an Erbb4-IR-dependent mechanism. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:180-190. [PMID: 37449045 PMCID: PMC10336735 DOI: 10.1016/j.omtn.2023.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023]
Abstract
Transforming growth factor β (TGF-β)/Smad3 plays a vital role in hypertensive cardiac fibrosis. The long non-coding RNA (lncRNA) Erbb4-IR is a novel Smad3-dependent lncRNA that mediates kidney fibrosis. However, the role of Erbb4-IR in hypertensive heart disease remains unexplored and was investigated in the present study by ultrasound-microbubble-mediated silencing of cardiac Erbb4-IR in hypertensive mice induced by angiotensin II. We found that chronic angiotensin II infusion induced hypertension and upregulated cardiac Erbb4-IR, which was associated with cardiac dysfunction, including a decrease in left ventricle ejection fraction (LVEF) and LV fractional shortening (LVFS) and an increase in LV mass. Knockdown of cardiac Erbb4-IR by Erbb4-IR short hairpin RNA (shRNA) gene transfer effectively improved the angiotensin II-induced deterioration of cardiac function, although blood pressure was not altered. Furthermore, silencing cardiac Erbb4-IR also inhibited angiotensin II-induced progressive cardiac fibrosis, as evidenced by reduced collagen I and III, alpha-smooth muscle actin (α-SMA), and fibronectin accumulation. Mechanistically, improved hypertensive cardiac injury by specifically silencing cardiac Erbb4-IR was associated with increased myocardial Smad7 and miR-29b, revealing that Erbb4-IR may target Smad7 and miR-29b to mediate angiotensin II-induced hypertensive cardiac fibrosis. In conclusion, Erbb4-IR is pathogenic in angiotensin II (Ang II)-induced cardiac remodeling, and targeting Erbb4-IR may be a novel therapy for hypertensive cardiovascular diseases.
Collapse
Affiliation(s)
- Jian-Chun Li
- Research Center of Integrated Traditional Chinese and Western Medicine, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jian Jia
- Research Center of Integrated Traditional Chinese and Western Medicine, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Li Dong
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- National Traditional Chinese Medicine Clinical Research Base, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhong-Jing Hu
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- National Traditional Chinese Medicine Clinical Research Base, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Hong-Lian Wang
- Research Center of Integrated Traditional Chinese and Western Medicine, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Li Wang
- Research Center of Integrated Traditional Chinese and Western Medicine, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Si-Jin Yang
- National Traditional Chinese Medicine Clinical Research Base, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Hui-Yao Lan
- Research Center of Integrated Traditional Chinese and Western Medicine, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| |
Collapse
|
4
|
Ou Y, Zong D, Ouyang R. Role of epigenetic abnormalities and intervention in obstructive sleep apnea target organs. Chin Med J (Engl) 2023; 136:631-644. [PMID: 35245923 PMCID: PMC10129098 DOI: 10.1097/cm9.0000000000002080] [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: 08/03/2022] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Obstructive sleep apnea (OSA) is a common condition that has considerable impacts on human health. Epigenetics has become a rapidly developing and exciting area in biology, and it is defined as heritable alterations in gene expression and has regulatory effects on disease progression. However, the published literature that is integrating both of them is not sufficient. The purpose of this article is to explore the relationship between OSA and epigenetics and to offer better diagnostic methods and treatment options. Epigenetic modifications mainly manifest as post-translational modifications in DNA and histone proteins and regulation of non-coding RNAs. Chronic intermittent hypoxia-mediated epigenetic alterations are involved in the progression of OSA and diverse multiorgan injuries, including cardiovascular disease, metabolic disorders, pulmonary hypertension, neural dysfunction, and even tumors. This article provides deeper insights into the disease mechanism of OSA and potential applications of targeted diagnosis, treatment, and prognosis in OSA complications.
Collapse
Affiliation(s)
- Yanru Ou
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Dandan Zong
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Ruoyun Ouyang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
5
|
The Function and Therapeutic Potential of lncRNAs in Cardiac Fibrosis. BIOLOGY 2023; 12:biology12020154. [PMID: 36829433 PMCID: PMC9952806 DOI: 10.3390/biology12020154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023]
Abstract
Cardiac fibrosis remains an unresolved problem in cardiovascular diseases. Fibrosis of the myocardium plays a key role in the clinical outcomes of patients with heart injuries. Moderate fibrosis is favorable for cardiac structure maintaining and contractile force transmission, whereas adverse fibrosis generally progresses to ventricular remodeling and cardiac systolic or diastolic dysfunction. The molecular mechanisms involved in these processes are multifactorial and complex. Several molecular mechanisms, such as TGF-β signaling pathway, extracellular matrix (ECM) synthesis and degradation, and non-coding RNAs, positively or negatively regulate myocardial fibrosis. Long noncoding RNAs (lncRNAs) have emerged as significant mediators in gene regulation in cardiovascular diseases. Recent studies have demonstrated that lncRNAs are crucial in genetic programming and gene expression during myocardial fibrosis. We summarize the function of lncRNAs in cardiac fibrosis and their contributions to miRNA expression, TGF-β signaling, and ECMs synthesis, with a particular attention on the exosome-derived lncRNAs in the regulation of adverse fibrosis as well as the mode of action of lncRNAs secreted into exosomes. We also discuss how the current knowledge on lncRNAs can be applied to develop novel therapeutic strategies to prevent or reverse cardiac fibrosis.
Collapse
|
6
|
Varghese LN, Schwenke DO, Katare R. Role of noncoding RNAs in cardiac ageing. Front Cardiovasc Med 2023; 10:1142575. [PMID: 37034355 PMCID: PMC10073704 DOI: 10.3389/fcvm.2023.1142575] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
The global population is estimated to reach 9.8 billion by 2050, of which 2.1 billion will comprise individuals above 60 years of age. As the number of elderly is estimated to double from 2017, it is a victory of the modern healthcare system but also worrisome as ageing, and the onset of chronic disease are correlated. Among other chronic conditions, cardiovascular diseases (CVDs) are the leading cause of death in the aged population. While the underlying cause of the age-associated development of CVDs is not fully understood, studies indicate the role of non-coding RNAs such as microRNAs (miRNAs) and long noncoding RNAs (lnc-RNAs) in the development of age-associated CVDs. miRNAs and lnc-RNAs are non-coding RNAs which control gene expression at the post-transcriptional level. The expression of specific miRNAs and lnc-RNAs are reportedly dysregulated with age, leading to cardiovascular system changes and ultimately causing CVDs. Since miRNAs and lnc-RNAs play several vital roles in maintaining the normal functioning of the cardiovascular system, they are also being explored for their therapeutic potential as a treatment for CVDs. This review will first explore the pathophysiological changes associated with ageing. Next, we will review the known mechanisms underlying the development of CVD in ageing with a specific focus on miRNA and lnc-RNAs. Finally, we will discuss the therapeutic options and future challenges towards healthy cardiac ageing. With the global ageing population on the rise, this review will provide a fundamental understanding of some of the underlying molecular mechanisms of cardiac ageing.
Collapse
|
7
|
Involvement of circRNAs in the Development of Heart Failure. Int J Mol Sci 2022; 23:ijms232214129. [PMID: 36430607 PMCID: PMC9697219 DOI: 10.3390/ijms232214129] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/05/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
In recent years, interest in non-coding RNAs as important physiological regulators has grown significantly. Their participation in the pathophysiology of cardiovascular diseases is extremely important. Circular RNA (circRNA) has been shown to be important in the development of heart failure. CircRNA is a closed circular structure of non-coding RNA fragments. They are formed in the nucleus, from where they are transported to the cytoplasm in a still unclear mechanism. They are mainly located in the cytoplasm or contained in exosomes. CircRNA expression varies according to the type of tissue. In the brain, almost 12% of genes produce circRNA, while in the heart it is only 9%. Recent studies indicate a key role of circRNA in cardiomyocyte hypertrophy, fibrosis, autophagy and apoptosis. CircRNAs act mainly by interacting with miRNAs through a "sponge effect" mechanism. The involvement of circRNA in the development of heart failure leads to the suggestion that they may be promising biomarkers and useful targets in the treatment of cardiovascular diseases. In this review, we will provide a brief introduction to circRNA and up-to-date understanding of their role in the mechanisms leading to the development of heart failure.
Collapse
|
8
|
Zheng X, Zhong T, Yu F, Duan J, Tang Y, Liu Y, Li M, Sun D, Yin D. Deficiency of a novel lncRNA-HRAT protects against myocardial ischemia reperfusion injury by targeting miR-370-3p/RNF41 pathway. Front Cardiovasc Med 2022; 9:951463. [PMID: 36172578 PMCID: PMC9510651 DOI: 10.3389/fcvm.2022.951463] [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: 05/24/2022] [Accepted: 08/22/2022] [Indexed: 01/17/2023] Open
Abstract
Accumulating evidence indicates that long non-coding RNAs (lncRNAs) contribute to myocardial ischemia/reperfusion (I/R) injury. However, the underlying mechanisms by which lncRNAs modulate myocardial I/R injury have not been thoroughly examined and require further investigation. A novel lncRNA named lncRNA-hypoxia/reoxygenation (H/R)-associated transcript (lncRNA-HRAT) was identified by RNA sequencing analysis. The expression of lncRNA-HRAT exhibited a significant increase in the I/R mice hearts and cardiomyocytes treated with H/R. LncRNA-HRAT overexpression facilitates H/R-induced cardiomyocyte apoptosis. Furthermore, cardiomyocyte-specific deficiency of lncRNA-HRAT in vivo after I/R decreased creatine kinase (CK) release in the serum, reduced myocardial infarct area, and improved cardiac dysfunction. Molecular mechanistic investigations revealed that lncRNA-HRAT serves as a competing endogenous RNA (ceRNA) of miR-370-3p, thus upregulating the expression of ring finger protein 41 (RNF41), thereby aggravating apoptosis in cardiomyocytes induced by H/R. This study revealed that the lncRNA-HRAT/miR-370-3p/RNF41 pathway regulates cardiomyocyte apoptosis and myocardial injury. These findings suggest that targeted inhibition of lncRNA-HRAT may offer a novel therapeutic method to prevent myocardial I/R injury.
Collapse
Affiliation(s)
- Xinbin Zheng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, China
| | - Ting Zhong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Fan Yu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Jingsi Duan
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Yao Tang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Yaxiu Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Mingrui Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Deqiang Sun
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Deling Yin
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
- *Correspondence: Deling Yin,
| |
Collapse
|
9
|
Chen G, Chen H, Zeng X, Zhu W. Stem cell-derived exosomal transcriptomes for wound healing. Front Surg 2022; 9:933781. [PMID: 36034367 PMCID: PMC9417542 DOI: 10.3389/fsurg.2022.933781] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/01/2022] [Indexed: 12/02/2022] Open
Abstract
Wound healing is a complex and integrated process of the interaction of various components within the injured tissue. Accumulating evidence suggested that stem cell-derived exosomal transcriptomes could serve as key regulatory molecules in wound healing in stem cell therapy. Stem cell-derived exosomal transcriptomes mainly consist of long noncoding RNAs (lncRNAs), microRNAs (miRNAs), circular RNAs (circRNAs) and messenger RNAs (mRNAs). In this article we presented a brief introduction on the wound repair process and exosomal transcriptomes. Meanwhile, we summarized our current knowledge of the involvement of exosomal transcriptomes in physiological and pathological wound repair process including inflammation, angiogenesis, and scar formation.
Collapse
Affiliation(s)
- Guiling Chen
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- National Institute of Stem Cell Clinical Research, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Hankun Chen
- Research and Development Department, Guangzhou Qinglan Biotechnology Company Limited, Guangzhou, China
| | - Xiang Zeng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- National Institute of Stem Cell Clinical Research, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Correspondence: Xiang Zeng Wei Zhu
| | - Wei Zhu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Correspondence: Xiang Zeng Wei Zhu
| |
Collapse
|
10
|
Zhao X, Ling F, Zhang GW, Yu N, Yang J, Xin XY. The Correlation Between MicroRNAs and Diabetic Retinopathy. Front Immunol 2022; 13:941982. [PMID: 35958584 PMCID: PMC9358975 DOI: 10.3389/fimmu.2022.941982] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/23/2022] [Indexed: 11/23/2022] Open
Abstract
Micro ribonucleic acids (miRNAs), as a category of post-transcriptional gene inhibitors, have a wide range of biological functions, are involved in many pathological processes, and are attractive therapeutic targets. Considerable evidence in ophthalmology indicates that miRNAs play an important role in diabetic retinopathy (DR), especially in inflammation, oxidative stress, and neurodegeneration. Targeting specific miRNAs for the treatment of DR has attracted much attention. This is a review focusing on the pathophysiological roles of miRNAs in DR, diabetic macular edema, and proliferative DR complex multifactorial retinal diseases, with particular emphasis on how miRNAs regulate complex molecular pathways and underlying pathomechanisms. Moreover, the future development potential and application limitations of therapy that targets specific miRNAs for DR are discussed.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Ophthalmology, Inner Mongolia Baogang Hospita, Baotou, Inner Mongolia, China
| | - Feng Ling
- Department of Ophthalmology, Inner Mongolia Baogang Hospita, Baotou, Inner Mongolia, China
| | - Guang wei Zhang
- Department of Cardiology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Na Yu
- Department of Scientific research, Inner Mongolia Baogang Hospita, Baotou, Inner Mongolia, China
| | - Jing Yang
- Department of Biology, Inner Mongolia University of Science and Technology Baotou Medical College, Baotou, Inner Mongolia, China
- *Correspondence: Jing Yang, ; Xiang yang Xin,
| | - Xiang yang Xin
- Department of Ophthalmology, Inner Mongolia Baogang Hospita, Baotou, Inner Mongolia, China
- *Correspondence: Jing Yang, ; Xiang yang Xin,
| |
Collapse
|
11
|
Hosseini M, Sahebi R, Aghasizadeh M, Yazdi DF, Salaribaghoonabad R, Godsi A, Soflaei SS, Mousavitaherabad SP, Ahmadihoseini A, Bajgiran MM, Ferns GA, Darban RA, Mobarhan MG. Investigating the predictive value of microRNA21 as a biomarker in induced myocardial infarction animal model. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
12
|
Correia CCM, Rodrigues LF, de Avila Pelozin BR, Oliveira EM, Fernandes T. Long Non-Coding RNAs in Cardiovascular Diseases: Potential Function as Biomarkers and Therapeutic Targets of Exercise Training. Noncoding RNA 2021; 7:ncrna7040065. [PMID: 34698215 PMCID: PMC8544698 DOI: 10.3390/ncrna7040065] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022] Open
Abstract
Despite advances in treatments and therapies, cardiovascular diseases (CVDs) remain one of the leading causes of death worldwide. The discovery that most of the human genome, although transcribed, does not encode proteins was crucial for focusing on the potential of long non-coding RNAs (lncRNAs) as essential regulators of cell function at the epigenetic, transcriptional, and post-transcriptional levels. This class of non-coding RNAs is related to the pathophysiology of the cardiovascular system. The different expression profiles of lncRNAs, in different contexts of CVDs, change a great potential in their use as a biomarker and targets of therapeutic intervention. Furthermore, regular physical exercise plays a protective role against CVDs; on the other hand, little is known about its underlying molecular mechanisms. In this review, we look at the accumulated knowledge on lncRNAs and their functions in the cardiovascular system, focusing on the cardiovascular pathology of arterial hypertension, coronary heart disease, acute myocardial infarction, and heart failure. We discuss the potential of these molecules as biomarkers for clinical use, their limitations, and how the manipulation of the expression profile of these transcripts through physical exercise can begin to be suggested as a strategy for the treatment of CVDs.
Collapse
Affiliation(s)
- Camila Caldas Martins Correia
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-030, Brazil;
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.d.A.P.); (E.M.O.)
| | - Luis Felipe Rodrigues
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.d.A.P.); (E.M.O.)
| | - Bruno Rocha de Avila Pelozin
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.d.A.P.); (E.M.O.)
| | - Edilamar Menezes Oliveira
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.d.A.P.); (E.M.O.)
| | - Tiago Fernandes
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.d.A.P.); (E.M.O.)
- Correspondence: ; Tel.: + 55-11-2648-1566 (ext. 05508-030)
| |
Collapse
|
13
|
Abstract
Cardiac hypertrophy, characterized by the enlargement of cardiomyocytes, is initially an adaptive response to physiological and pathological stimuli. Decompensated cardiac hypertrophy is related to fibrosis, inflammatory cytokine, maladaptive remodeling, and heart failure. Although pathological myocardial hypertrophy is the main cause of hypertrophy-related morbidity and mortality, our understanding of its mechanism is still poor. Long noncoding RNAs (lncRNAs) are noncoding RNAs that regulate various physiological and pathological processes through multiple molecular mechanisms. Recently, accumulating evidence has indicated that lncRNA-H19 is a potent regulator of the progression of cardiac hypertrophy. For the first time, this review summarizes the current studies about the role of lncRNA-H19 in cardiac hypertrophy, including its pathophysiological processes and underlying pathological mechanism, including calcium regulation, fibrosis, apoptosis, angiogenesis, inflammation, and methylation. The context within which lncRNA-H19 might be developed as a target for cardiac hypertrophy treatment is then discussed to gain better insight into the possible biological functions of lncRNA-H19 in cardiac hypertrophy.
Collapse
|
14
|
Wang W, Min L, Qiu X, Wu X, Liu C, Ma J, Zhang D, Zhu L. Biological Function of Long Non-coding RNA (LncRNA) Xist. Front Cell Dev Biol 2021; 9:645647. [PMID: 34178980 PMCID: PMC8222981 DOI: 10.3389/fcell.2021.645647] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) regulate gene expression in a variety of ways at epigenetic, chromatin remodeling, transcriptional, and translational levels. Accumulating evidence suggests that lncRNA X-inactive specific transcript (lncRNA Xist) serves as an important regulator of cell growth and development. Despites its original roles in X-chromosome dosage compensation, lncRNA Xist also participates in the development of tumor and other human diseases by functioning as a competing endogenous RNA (ceRNA). In this review, we comprehensively summarized recent progress in understanding the cellular functions of lncRNA Xist in mammalian cells and discussed current knowledge regarding the ceRNA network of lncRNA Xist in various diseases. Long non-coding RNAs (lncRNAs) are transcripts that are more than 200 nt in length and without an apparent protein-coding capacity (Furlan and Rougeulle, 2016; Maduro et al., 2016). These RNAs are believed to be transcribed by the approximately 98-99% non-coding regions of the human genome (Derrien et al., 2012; Fu, 2014; Montalbano et al., 2017; Slack and Chinnaiyan, 2019), as well as a large variety of genomic regions, such as exonic, tronic, and intergenic regions. Hence, lncRNAs are also divided into eight categories: Intergenic lncRNAs, Intronic lncRNAs, Enhancer lncRNAs, Promoter lncRNAs, Natural antisense/sense lncRNAs, Small nucleolar RNA-ended lncRNAs (sno-lncRNAs), Bidirectional lncRNAs, and non-poly(A) lncRNAs (Ma et al., 2013; Devaux et al., 2015; St Laurent et al., 2015; Chen, 2016; Quinn and Chang, 2016; Richard and Eichhorn, 2018; Connerty et al., 2020). A range of evidence has suggested that lncRNAs function as key regulators in crucial cellular functions, including proliferation, differentiation, apoptosis, migration, and invasion, by regulating the expression level of target genes via epigenomic, transcriptional, or post-transcriptional approaches (Cao et al., 2018). Moreover, lncRNAs detected in body fluids were also believed to serve as potential biomarkers for the diagnosis, prognosis, and monitoring of disease progression, and act as novel and potential drug targets for therapeutic exploitation in human disease (Jiang W. et al., 2018; Zhou et al., 2019a). Long non-coding RNA X-inactive specific transcript (lncRNA Xist) are a set of 15,000-20,000 nt sequences localized in the X chromosome inactivation center (XIC) of chromosome Xq13.2 (Brown et al., 1992; Debrand et al., 1998; Kay, 1998; Lee et al., 2013; da Rocha and Heard, 2017; Yang Z. et al., 2018; Brockdorff, 2019). Previous studies have indicated that lncRNA Xist regulate X chromosome inactivation (XCI), resulting in the inheritable silencing of one of the X-chromosomes during female cell development. Also, it serves a vital regulatory function in the whole spectrum of human disease (notably cancer) and can be used as a novel diagnostic and prognostic biomarker and as a potential therapeutic target for human disease in the clinic (Liu et al., 2018b; Deng et al., 2019; Dinescu et al., 2019; Mutzel and Schulz, 2020; Patrat et al., 2020; Wang et al., 2020a). In particular, lncRNA Xist have been demonstrated to be involved in the development of multiple types of tumors including brain tumor, Leukemia, lung cancer, breast cancer, and liver cancer, with the prominent examples outlined in Table 1. It was also believed that lncRNA Xist (Chaligne and Heard, 2014; Yang Z. et al., 2018) contributed to other diseases, such as pulmonary fibrosis, inflammation, neuropathic pain, cardiomyocyte hypertrophy, and osteoarthritis chondrocytes, and more specific details can be found in Table 2. This review summarizes the current knowledge on the regulatory mechanisms of lncRNA Xist on both chromosome dosage compensation and pathogenesis (especially cancer) processes, with a focus on the regulatory network of lncRNA Xist in human disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dongyi Zhang
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, China
| | - Lingyun Zhu
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, China
| |
Collapse
|
15
|
Giordo R, Ahmed YMA, Allam H, Abusnana S, Pappalardo L, Nasrallah GK, Mangoni AA, Pintus G. EndMT Regulation by Small RNAs in Diabetes-Associated Fibrotic Conditions: Potential Link With Oxidative Stress. Front Cell Dev Biol 2021; 9:683594. [PMID: 34095153 PMCID: PMC8170089 DOI: 10.3389/fcell.2021.683594] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Diabetes-associated complications, such as retinopathy, nephropathy, cardiomyopathy, and atherosclerosis, the main consequences of long-term hyperglycemia, often lead to organ dysfunction, disability, and increased mortality. A common denominator of these complications is the myofibroblast-driven excessive deposition of extracellular matrix proteins. Although fibroblast appears to be the primary source of myofibroblasts, other cells, including endothelial cells, can generate myofibroblasts through a process known as endothelial to mesenchymal transition (EndMT). During EndMT, endothelial cells lose their typical phenotype to acquire mesenchymal features, characterized by the development of invasive and migratory abilities as well as the expression of typical mesenchymal products such as α-smooth muscle actin and type I collagen. EndMT is involved in many chronic and fibrotic diseases and appears to be regulated by complex molecular mechanisms and different signaling pathways. Recent evidence suggests that small RNAs, in particular microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), are crucial mediators of EndMT. Furthermore, EndMT and miRNAs are both affected by oxidative stress, another key player in the pathophysiology of diabetic fibrotic complications. In this review, we provide an overview of the primary redox signals underpinning the diabetic-associated fibrotic process. Then, we discuss the current knowledge on the role of small RNAs in the regulation of EndMT in diabetic retinopathy, nephropathy, cardiomyopathy, and atherosclerosis and highlight potential links between oxidative stress and the dyad small RNAs-EndMT in driving these pathological states.
Collapse
Affiliation(s)
- Roberta Giordo
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Yusra M. A. Ahmed
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Hilda Allam
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Salah Abusnana
- Department of Diabetes and Endocrinology, University Hospital Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Lucia Pappalardo
- Department of Biology, Chemistry and Environmental Studies, American University of Sharjah, Sharjah, United Arab Emirates
| | - Gheyath K. Nasrallah
- Department of Biomedical Sciences, College of Health Sciences Member of QU Health, Qatar University, Doha, Qatar
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Arduino Aleksander Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Flinders Medical Centre, Adelaide, SA, Australia
| | - Gianfranco Pintus
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
16
|
Wang JF, Xi ZN, Su HJ, Bao Z, Qiao YH. SP1-induced overexpression of LINC00520 facilitates non-small cell lung cancer progression through miR-577/CCNE2 pathway and predicts poor prognosis. Hum Cell 2021; 34:952-964. [PMID: 33728585 DOI: 10.1007/s13577-021-00518-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/05/2021] [Indexed: 01/13/2023]
Abstract
Long noncoding RNAs (lncRNAs) have gained much attention in the past few years. Long intergenic non-protein coding RNA 520 (LINC00520) was one of the newly discovered lncRNA which has been demonstrated to be dysregulated in several cancers. So far, the function and mechanism of LINC00520 in non-small cell lung cancer (NSCLC) are unclear. In this paper, our group first showed that LINC00520 level was elevated in non-small cell lung cancer (NSCLC) tissue and cells. In addition, SP1 could bind directly to the promoter region of LINC00520 and thus promote its transcription. Increased LINC00520 was distinctly correlated with advanced tumor stage and shorter survival time in NSCLC patients. Further functional investigations provided evidences that forced down regulation of LINC00520 inhibited NSCLC cell proliferation, invasion, metastasis and EMT, while contributing to cells apoptosis. Mechanistically, we found that LINC00520 serving as a competing endogenous RNA to be involved in the modulation of miR-577 expressions, and thus affected the expression of CCNE2 which was a target gene of miR-577. Moreover, in NSCLC cells with si-LINC00520, up regulation of CCNE2 led to an increase of cell growth and invasion. Taken together, LINC00520 displayed its tumor-promotive roles through modulating the miR-577/CCNE2 axis, highlighting a potential therapeutic strategy for NSCLC patients.
Collapse
Affiliation(s)
- Ji-Fang Wang
- Department of Respiration Ward II, Henan Provincial Chest Hospital, Weiwu Road No. 1, Zhengzhou, Henan, China
| | - Zhuo-Na Xi
- Department of Respiration Ward II, Henan Provincial Chest Hospital, Weiwu Road No. 1, Zhengzhou, Henan, China
| | - Hong-Jian Su
- Department of Respiration Ward II, Henan Provincial Chest Hospital, Weiwu Road No. 1, Zhengzhou, Henan, China
| | - Zhen Bao
- Department of Respiration Ward II, Henan Provincial Chest Hospital, Weiwu Road No. 1, Zhengzhou, Henan, China
| | - Ya-Hong Qiao
- Department of Respiration Ward II, Henan Provincial Chest Hospital, Weiwu Road No. 1, Zhengzhou, Henan, China.
| |
Collapse
|
17
|
Cao X, Ma Q, Wang B, Qian Q, Liu N, Liu T, Dong X. Silencing long non-coding RNA MIAT ameliorates myocardial dysfunction induced by myocardial infarction via MIAT/miR-10a-5p/EGR2 axis. Aging (Albany NY) 2021; 13:11188-11206. [PMID: 33819189 PMCID: PMC8109106 DOI: 10.18632/aging.202785] [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: 02/13/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
Long non-coding RNA (lncRNA) myocardial infarction-associated transcript (MIAT) has been widely-demonstrated to function as diagnostic markers for acute myocardial infarction (MI). This study was designed to explore the modulatory role of MIAT and its underlying molecular mechanism in MI. Firstly, MI mouse model was developed via ligation of the descending branch of the left coronary artery, and cell model was established through exposure to hypoxic conditions. Online prediction indicated that MIAT could bind to microRNA-10a-5p (miR-10a-5p), while miR-10a-5p was highlighted to bind to early growth response gene-2 (EGR2). MIAT and EGR2 were subsequently determined to be highly-expressed, whereas miR-10a-5p was found to be poorly-expressed in cardiomyocytes exposed to hypoxia as well as in MI mice using RT-qPCR and Western blot assay. The binding relationships between MIAT and miR-10a-5p, and between miR-10a-5p and EGR2 were further confirmed by dual-luciferase reporter and RNA immunoprecipitation assays. The results of in vitro and in vivo experimentation also suggested that overexpression of miR-10a-5p or silencing of MIAT and EGR2 reduced cardiomyocyte apoptosis and increased ATP content, thus alleviating the impairment of cardiac function following MI. In a word, inhibition of MIAT protects against cardiac dysfunction induced by MI through the crosstalk with miR-10a-5p/EGR2.
Collapse
Affiliation(s)
- Xiangke Cao
- School of Life Sciences, North China University of Science and Technology, Tangshan 063210, P.R. China
| | - Qinghua Ma
- Department of Preventive Health, The Third People's Hospital Of Xiangcheng District In Suzhou, Suzhou 215134, P.R. China
| | - Bin Wang
- Department of Pediatrics, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, P.R. China
| | - Qingqiang Qian
- Department of Neurology, Tangshan Gongren Hospital, Tangshan 063000, P.R. China
| | - Ning Liu
- Department of Cardiovascular Diseases, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, P.R. China
| | - Tiejun Liu
- Department of Anesthesiology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, P.R. China
| | - Xiaoliu Dong
- Department of Neurology, Tangshan People's Hospital, Tangshan 063001, P.R. China
| |
Collapse
|
18
|
Long Non-Coding RNAs (lncRNAs) in Cardiovascular Disease Complication of Type 2 Diabetes. Diagnostics (Basel) 2021; 11:diagnostics11010145. [PMID: 33478141 PMCID: PMC7835902 DOI: 10.3390/diagnostics11010145] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
The discovery of non-coding RNAs (ncRNAs) has opened a new paradigm to use ncRNAs as biomarkers to detect disease progression. Long non-coding RNAs (lncRNA) have garnered the most attention due to their specific cell-origin and their existence in biological fluids. Type 2 diabetes patients will develop cardiovascular disease (CVD) complications, and CVD remains the top risk factor for mortality. Understanding the lncRNA roles in T2D and CVD conditions will allow the future use of lncRNAs to detect CVD complications before the symptoms appear. This review aimed to discuss the roles of lncRNAs in T2D and CVD conditions and their diagnostic potential as molecular biomarkers for CVD complications in T2D.
Collapse
|
19
|
Luo B, He Z, Huang S, Wang J, Han D, Xue H, Liu P, Zeng X, Lu D. Long Non-Coding RNA 554 Promotes Cardiac Fibrosis via TGF-β1 Pathway in Mice Following Myocardial Infarction. Front Pharmacol 2020; 11:585680. [PMID: 33390954 PMCID: PMC7772239 DOI: 10.3389/fphar.2020.585680] [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: 07/21/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022] Open
Abstract
Rationale: Cardiac fibrosis is observed in nearly every form of myocardial disease. Long non-coding RNAs (lncRNAs) have been shown to play an important role in cardiac fibrosis, but the detailed molecular mechanism remains unknown. Object: We aimed at characterizing lncRNA 554 expression in murine cardiac fibroblasts (CFs) after myocardial infarction (MI) to identify CF-enriched lncRNA and investigate its function and contribution to cardiac fibrosis and function. Methods and Results: In this study, we identified lncRNA NONMMUT022554 (lncRNA 554) as a regulator of MI-induced cardiac fibrosis. We found that lncRNA 554 was significantly up-regulated in the mouse hearts following MI. Further study showed that lncRNA 554 was predominantly expressed in cardiac fibroblasts, indicating a potential role of lncRNA 554 in cardiac fibrosis. In vitro knockdown of lncRNA 554 by siRNA suppressed fibroblasts migration and expression of extracellular matrix (ECM); while overexpression of lncRNA 554 promoted expression of ECM genes. Consistently, lentivirus mediated in vivo knockdown of lncRNA 554 could inhibit cardiac fibrosis and improve cardiac function in mouse model of MI. More importantly, TGF-β1 inhibitor (TEW-7197) could reverse the pro-fibrotic function of lncRNA 554 in CFs. This suggests that the effects of lncRNA 554 on cardiac fibrosis is TGF-β1 dependent. Conclusion: Collectively, our study illustrated the role of lncRNA 554 in cardiac fibrosis, suggested that lncRNA 554 might be a novel target for cardiac fibrosis.
Collapse
Affiliation(s)
- Bihui Luo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| | - Zhiyu He
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| | - Shijun Huang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, China.,Department of Cardiovascular Medicine, Yue Bei People's Hospital, Guangdong, China
| | - Jinping Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| | - Dunzheng Han
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| | - Hao Xue
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| | - Peiying Liu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| | - Xiaojun Zeng
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| | - Dongfeng Lu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| |
Collapse
|
20
|
Chen YQ, Yang X, Xu W, Yan Y, Chen XM, Huang ZQ. Knockdown of lncRNA TTTY15 alleviates myocardial ischemia-reperfusion injury through the miR-374a-5p/FOXO1 axis. IUBMB Life 2020; 73:273-285. [PMID: 33296140 DOI: 10.1002/iub.2428] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/13/2020] [Accepted: 11/30/2020] [Indexed: 01/11/2023]
Abstract
Myocardial ischemia/reperfusion (I/R) injury greatly contributes to myocardial tissue damage in patients with coronary disease, which eventually leads to heart failure. Long noncoding RNAs (lncRNAs) have an emerging role in the process of myocardial I/R injury. Our previous work revealed the protective role of miR-374a-5p against myocardial I/R injury. In this study, we explored the role of lncRNA TTTY15 and its potential interaction mechanisms with miR-374a-5p in myocardial I/R injury. The expression of TTTY15 was increased both in vitro and in vivo after myocardial I/R injury models according to quantitative real-time polymerase chain reaction. Various assays were conducted to evaluate the regulatory relationship among TTTY15, miR-374a-5p, FOXO1, and autophagy in H9c2 and HL-1 cells. The results showed that TTTY15 suppresses autophagy and myocardial I/R injury by targeting miR-374a-5p. We found that TTTY15 regulates miR-374a-5p, thus affecting FOXO1 expression and autophagy in myocytes during I/R. Furthermore, in an in vivo mouse model of myocardial I/R injury, suppression of TTTY15 successfully alleviated myocardial I/R injury. Our results reveal a novel feedback mechanism in which TTTY15 regulates miRNA processing and a potential target in myocardial I/R injury. TTTY15 is a promising therapeutic target for treating myocardial I/R injury.
Collapse
Affiliation(s)
- Yong-Quan Chen
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Xin Yang
- Department of Geriatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Wei Xu
- Department of Cardiology, Huadu District People's Hospital of Guangzhou, Guangzhou, PR China
| | - Yi Yan
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Xi-Ming Chen
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Zhao-Qi Huang
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| |
Collapse
|
21
|
Greco S, Madè A, Gaetano C, Devaux Y, Emanueli C, Martelli F. Noncoding RNAs implication in cardiovascular diseases in the COVID-19 era. J Transl Med 2020; 18:408. [PMID: 33129318 PMCID: PMC7602761 DOI: 10.1186/s12967-020-02582-8] [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] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/24/2020] [Indexed: 12/21/2022] Open
Abstract
COronaVIrus Disease 19 (COVID-19) is caused by the infection of the Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2). Although the main clinical manifestations of COVID-19 are respiratory, many patients also display acute myocardial injury and chronic damage to the cardiovascular system. Understanding both direct and indirect damage caused to the heart and the vascular system by SARS-CoV-2 infection is necessary to identify optimal clinical care strategies. The homeostasis of the cardiovascular system requires a tight regulation of the gene expression, which is controlled by multiple types of RNA molecules, including RNA encoding proteins (messenger RNAs) (mRNAs) and those lacking protein-coding potential, the noncoding-RNAs. In the last few years, dysregulation of noncoding-RNAs has emerged as a crucial component in the pathophysiology of virtually all cardiovascular diseases. Here we will discuss the potential role of noncoding RNAs in COVID-19 disease mechanisms and their possible use as biomarkers of clinical use.
Collapse
Affiliation(s)
- S Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097, Milan, Italy
| | - A Madè
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097, Milan, Italy
| | - C Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | - Y Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, Strassen, Luxembourg
| | - C Emanueli
- Imperial College London, National Heart and Lung Institute, Hammersmith Campus, London, W12 0NN, UK
| | - F Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097, Milan, Italy.
| |
Collapse
|
22
|
Abstract
BACKGROUND The long noncoding RNAs have gradually been reported to be an important class of RNAs with pivotal roles in the development and progression of myocardial infarction (MI). In this study, we hypothesized that genetic variant of cyclin-dependent kinase inhibitor 2B antisense RNA (ANRIL) may affect the prognosis of MI patients. METHODS A systematic review and meta-analysis of studies including 11,269 cases and 10,707 controls on the association of 5 ANRIL single nucleotide polymorphism and the overall risk of MI or coronary artery disease (CAD) was performed. RESULTS In the meta-analysis, rs4977574 A > G, rs1333040 C > T, rs1333042 A > G and rs10757274 A > G ANRIL polymorphisms were correlated with overall MI or CAD risk. No significant associations were found between ANRIL rs1333049 G > C polymorphism and CAD risk. CONCLUSIONS The results indicated that ANRIL polymorphism (rs4977574, rs1333040, rs1333042, and rs10757274) were more generally associated with CAD or MI risk. Further experimental studies to evaluate the limits of this hypothesis are warranted, and future functional studies are required to clarify the possible mechanisms.
Collapse
Affiliation(s)
- Ya-nan Zhang
- Heilongjiang Provincial Hospital Affiliated to Harbin Institute of Technology
| | - Bo Qiang
- Department of orthopedics, Harbin fifth hospital, Harbin
| | - Li-juan Fu
- Department of infectious medicine, xiang ’an hospital of xiamen university, Xiamen, China
| |
Collapse
|
23
|
Lumley AI, Zhang L, Ernens I, Leszek P, Devaux Y. The Long Noncoding RNA Landscape of Cardiac Regeneration in Zebrafish. Can J Cardiol 2020; 37:484-492. [PMID: 32835855 DOI: 10.1016/j.cjca.2020.08.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/24/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Novel therapeutic targets of heart failure (HF) are needed. Long noncoding RNAs (lncRNAs) are engaged during cardiac regeneration. Unlike in humans, zebrafish naturally undergo cardiac regeneration after HF. We aimed to describe the landscape of lncRNAs during regeneration in a zebrafish model of HF and to investigate their human homologs. METHODS HF was established in adult zebrafish through thrice-weekly incubations with an anemia-inducing drug, phenylhydrazine hydrochloride (PHZ). After 5 weeks, PHZ treatment ceased and the fish were followed through a regeneration period of 14 days. Total RNA was extracted from the hearts of adult zebrafish after establishment of HF and at 2, 5, and 9 days after treatment cessation (9 hearts per condition at each time point). Gene regulation patterns were characterized with the use of bioinformatics and validated with the use of quantitative polymerase chain reaction. RESULTS We obtained 14,340 lncRNAs from the reannotated Affymetrix zebrafish microarray. Of these, 187 lncRNAs were found to be differentially expressed (false discovery rate < 0.05 and fold change ≥ 2) at at least 1 time point. 85% of differentially expressed lncRNAs overlapped or were close to (distance < 10 kb) protein-coding genes which were mostly related to muscle development in Gene Ontology analyses. Fifty-seven lncRNAs had human homologs, according to orientation relative to their conserved protein-coding neighbours. CONCLUSIONS LncRNAs are differentially expressed during regeneration after HF in adult zebrafish and could be potential future therapeutic targets. The extent to which lncRNAs contribute to cardiac regeneration is a worthy avenue for future research.
Collapse
Affiliation(s)
- Andrew I Lumley
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Lu Zhang
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Isabelle Ernens
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Przemyslaw Leszek
- Heart Failure and Transplantology Department, Institute of Cardiology, Warsaw, Poland
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg, Luxembourg.
| |
Collapse
|
24
|
Gu S, Dai J, Qu T, He Z. Emerging Roles of MicroRNAs and Long Noncoding RNAs in Cadmium Toxicity. Biol Trace Elem Res 2020; 195:481-490. [PMID: 31422539 DOI: 10.1007/s12011-019-01859-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023]
Abstract
Metal cadmium (Cd) and its compounds are ubiquitous industrial and environmental pollutants and they have been believed to exert severe damage to multiple organs and tissues. MicroRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are the two most common noncoding RNAs and have pivotal roles in various cellular and physiological processes. Since the importance of miRNAs and lncRNAs in Cd toxicity has been widely recognized, we focus our interests on the current researches of miRNAs and lncRNAs as well as their regulation roles in Cd toxicity. In this paper, the keywords "cadmium" in combination with "miRNA" or "LncRNA" or "noncoding RNA" was used to retrieve relevant articles in PubMed, EMbase, CNKI, Wan Fang, and CBM databases. The literatures which contained the above keywords and carried out in animals (in vivo and in vitro) have been collected, collated, analyzed, and summarized. Our summary results showed that hundreds of miRNAs and lncRNAs are involved in the Cd toxicity, which have been demonstrated as multiple organ injury, reproductive toxicity, malignant transformation, and abnormal repair of DNA damage. In this paper, we also discussed the blank in present research field of Cd toxicity as well as suggested some ideas for future study in Cd toxicity.
Collapse
Affiliation(s)
- Shiyan Gu
- Department of Environmental and Occupational Health, Faculty of Public Health, Dali University, No. 22, Wanhua Road, Dali, 671000, Yunnan, People's Republic of China.
| | - Jiao Dai
- Qujing Medical College, Qujing, Yunnan, China
| | - Tengjiao Qu
- Department of Environmental and Occupational Health, Faculty of Public Health, Dali University, No. 22, Wanhua Road, Dali, 671000, Yunnan, People's Republic of China
| | - Zuoshun He
- Department of Environmental and Occupational Health, Faculty of Public Health, Dali University, No. 22, Wanhua Road, Dali, 671000, Yunnan, People's Republic of China.
| |
Collapse
|
25
|
RNAs in Brain and Heart Diseases. Int J Mol Sci 2020; 21:ijms21103717. [PMID: 32466222 PMCID: PMC7279324 DOI: 10.3390/ijms21103717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
In the era of single-cell analysis, one always has to keep in mind the systemic nature of various diseases and how these diseases could be optimally studied. Comorbidities of the heart in neurological diseases as well as of the brain in cardiovascular diseases are prevalent, but how interactions in the brain–heart axis affect disease development and progression has been poorly addressed. Several brain and heart diseases share common risk factors. A better understanding of the brain–heart interactions will provide better insights for future treatment and personalization of healthcare, for heart failure patients’ benefit notably. We review here emerging evidence that studying noncoding RNAs in the brain–heart axis could be pivotal in understanding these interactions. We also introduce the Special Issue of the International Journal of Molecular Sciences RNAs in Brain and Heart Diseases—EU-CardioRNA COST Action.
Collapse
|
26
|
Muñoz P, Ardiles ÁO, Pérez-Espinosa B, Núñez-Espinosa C, Paula-Lima A, González-Billault C, Espinosa-Parrilla Y. Redox modifications in synaptic components as biomarkers of cognitive status, in brain aging and disease. Mech Ageing Dev 2020; 189:111250. [PMID: 32433996 DOI: 10.1016/j.mad.2020.111250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/05/2020] [Accepted: 04/10/2020] [Indexed: 02/06/2023]
Abstract
Aging is a natural process that includes several changes that gradually make organisms degenerate and die. Harman's theory proposes that aging is a consequence of the progressive accumulation of oxidative modifications mediated by reactive oxygen/nitrogen species, which plays an essential role in the development and progression of many neurodegenerative diseases. This review will focus on how abnormal redox modifications induced by age impair the functionality of neuronal redox-sensitive proteins involved in axonal elongation and guidance, synaptic plasticity, and intercellular communication. We will discuss post-transcriptional regulation of gene expression by microRNAs as a mechanism that controls the neuronal redox state. Finally, we will discuss how some brain-permeant antioxidants from the diet have a beneficial effect on cognition. Taken together, the evidence revised here indicates that oxidative-driven modifications of specific proteins and changes in microRNA expression may be useful biomarkers for aging and neurodegenerative diseases. Also, some specific antioxidant therapies have undoubtedly beneficial neuroprotective effects when administered in the correct doses, in the ideal formulation combination, and during the appropriate therapeutic window. The use of some antioxidants is, therefore, still poorly explored for the treatment of neurodegenerative diseases such as Alzheimer's disease.
Collapse
Affiliation(s)
- Pablo Muñoz
- Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile; Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile; Biomedical Research Center, Universidad de Valparaíso, Valparaíso, Chile; Thematic Task Force on Healthy Aging, CUECH Research Network.
| | - Álvaro O Ardiles
- Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile; Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile; Thematic Task Force on Healthy Aging, CUECH Research Network; Interdisciplinary Center of Neuroscience of Valparaíso, Universidad de Valparaíso, Valparaíso, Chile; Interdisciplinary Center for Health Studies, Universidad de Valparaíso, Valparaíso, Chile
| | - Boris Pérez-Espinosa
- Thematic Task Force on Healthy Aging, CUECH Research Network; Laboratorio biología de la Reproduccion, Departamento Biomédico, Facultad Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Cristian Núñez-Espinosa
- Thematic Task Force on Healthy Aging, CUECH Research Network; School of Medicine, Universidad de Magallanes, Punta Arenas, Chile
| | - Andrea Paula-Lima
- Thematic Task Force on Healthy Aging, CUECH Research Network; Institute for Research in Dental Sciences, Faculty of Dentistry; Universidad de Chile, Santiago, Chile; Biomedical Neuroscience Institute (BNI) and Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Christian González-Billault
- Thematic Task Force on Healthy Aging, CUECH Research Network; Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; FONDAP Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, USA.
| | - Yolanda Espinosa-Parrilla
- Thematic Task Force on Healthy Aging, CUECH Research Network; School of Medicine, Universidad de Magallanes, Punta Arenas, Chile; Laboratory of Molecular Medicine - LMM, Center for Education, Healthcare and Investigation - CADI, University of Magallanes, Punta Arenas, Chile.
| |
Collapse
|
27
|
Lin B, Xu J, Wang F, Wang J, Zhao H, Feng D. LncRNA XIST promotes myocardial infarction by regulating FOS through targeting miR-101a-3p. Aging (Albany NY) 2020; 12:7232-7247. [PMID: 32315985 PMCID: PMC7202499 DOI: 10.18632/aging.103072] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 03/24/2020] [Indexed: 12/14/2022]
Abstract
The purpose of this study was to reveal the hypothesis that lncRNA X inactive specific transcript (XIST) can participate in the regulation of cardiomyocyte apoptosis in neonatal mice cardiomyocytes (NMCMs) and myocardial infarction (MI) through targeting miR-101a-3p. NMCMs were isolated from neonatal C57BL/6 mice and anoxia was induced in hypoxic chamber. MTT assay and flow cytometry were used to determine proliferation and apoptosis respectively. The target relationship among XIST, miR-101a-3p and FOS was revealed by bioinformatic analysis, luciferase reporter assay, pull-down assay and RNA immunoprecipitation assay. The expression of XIST, miR-101a-3p, FOS and apoptosis-related proteins was determined by qRT-PCR or western blot. MI model was constructed to reveal the role of XIST. We found that XIST was up-regulated in NMCMs under anoxia condition. Moreover, XIST increased FOS expression by sponging miR-101a-3p in anoxia cells. Silencing XIST expression improved cell viability and suppressed apoptosis in vitro and inhibited myocardial infarction by reducing the level of c-FOS and apoptosis-related proteins in vivo. Our findings suggest that XIST is involved in MI, modulation of its level can be used as a new strategy or potential target in the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Bin Lin
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jing Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Feng Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jiaxiang Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hui Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Deguang Feng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| |
Collapse
|
28
|
Valkov N, Das S. Y RNAs: Biogenesis, Function and Implications for the Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:327-342. [PMID: 32285422 DOI: 10.1007/978-981-15-1671-9_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In recent years, progress in the field of high-throughput sequencing technology and its application to a wide variety of biological specimens has greatly advanced the discovery and cataloging of a diverse set of non-coding RNAs (ncRNAs) that have been found to have unexpected biological functions. Y RNAs are an emerging class of highly conserved, small ncRNAs. There is a growing number of reports in the literature demonstrating that Y RNAs and their fragments are not just random degradation products but are themselves bioactive molecules. This review will outline what is currently known about Y RNA including biogenesis, structure and functional roles. In addition, we will provide an overview of studies reporting the presence and functions attributed to Y RNAs in the cardiovascular system.
Collapse
Affiliation(s)
- Nedyalka Valkov
- Cardiovascular Research Center of Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Saumya Das
- Cardiovascular Research Center of Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Kim S, Song J, Ernst P, Latimer MN, Ha CM, Goh KY, Ma W, Rajasekaran NS, Zhang J, Liu X, Prabhu SD, Qin G, Wende AR, Young ME, Zhou L. MitoQ regulates redox-related noncoding RNAs to preserve mitochondrial network integrity in pressure-overload heart failure. Am J Physiol Heart Circ Physiol 2020; 318:H682-H695. [PMID: 32004065 PMCID: PMC7099446 DOI: 10.1152/ajpheart.00617.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 01/04/2023]
Abstract
Evidence suggests that mitochondrial network integrity is impaired in cardiomyocytes from failing hearts. While oxidative stress has been implicated in heart failure (HF)-associated mitochondrial remodeling, the effect of mitochondrial-targeted antioxidants, such as mitoquinone (MitoQ), on the mitochondrial network in a model of HF (e.g., pressure overload) has not been demonstrated. Furthermore, the mechanism of this regulation is not completely understood with an emerging role for posttranscriptional regulation via long noncoding RNAs (lncRNAs). We hypothesized that MitoQ preserves mitochondrial fusion proteins (i.e., mitofusin), likely through redox-sensitive lncRNAs, leading to improved mitochondrial network integrity in failing hearts. To test this hypothesis, 8-wk-old C57BL/6J mice were subjected to ascending aortic constriction (AAC), which caused substantial left ventricular (LV) chamber remodeling and remarkable contractile dysfunction in 1 wk. Transmission electron microscopy and immunostaining revealed defective intermitochondrial and mitochondrial-sarcoplasmic reticulum ultrastructure in AAC mice compared with sham-operated animals, which was accompanied by elevated oxidative stress and suppressed mitofusin (i.e., Mfn1 and Mfn2) expression. MitoQ (1.36 mg·day-1·mouse-1, 7 consecutive days) significantly ameliorated LV dysfunction, attenuated Mfn2 downregulation, improved interorganellar contact, and increased metabolism-related gene expression. Moreover, our data revealed that MitoQ alleviated the dysregulation of an Mfn2-associated lncRNA (i.e., Plscr4). In summary, the present study supports a unique mechanism by which MitoQ improves myocardial intermitochondrial and mitochondrial-sarcoplasmic reticulum (SR) ultrastructural remodeling in HF by maintaining Mfn2 expression via regulation by an lncRNA. These findings underscore the important role of lncRNAs in the pathogenesis of HF and the potential of targeting them for effective HF treatment.NEW & NOTEWORTHY We have shown that MitoQ improves cardiac mitochondrial network integrity and mitochondrial-SR alignment in a pressure-overload mouse heart-failure model. This may be occurring partly through preventing the dysregulation of a redox-sensitive lncRNA-microRNA pair (i.e., Plscr4-miR-214) that results in an increase in mitofusin-2 expression.
Collapse
Affiliation(s)
- Seulhee Kim
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jiajia Song
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Patrick Ernst
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mary N Latimer
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Chae-Myeong Ha
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kah Yong Goh
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wenxia Ma
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Xiaoguang Liu
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sumanth D Prabhu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gangjian Qin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Adam R Wende
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Martin E Young
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lufang Zhou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
30
|
Kura B, Kalocayova B, Devaux Y, Bartekova M. Potential Clinical Implications of miR-1 and miR-21 in Heart Disease and Cardioprotection. Int J Mol Sci 2020; 21:ijms21030700. [PMID: 31973111 PMCID: PMC7037063 DOI: 10.3390/ijms21030700] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/10/2020] [Accepted: 01/17/2020] [Indexed: 02/07/2023] Open
Abstract
The interest in non-coding RNAs, which started more than a decade ago, has still not weakened. A wealth of experimental and clinical studies has suggested the potential of non-coding RNAs, especially the short-sized microRNAs (miRs), to be used as the new generation of therapeutic targets and biomarkers of cardiovascular disease, an ever-growing public health issue in the modern world. Among the hundreds of miRs characterized so far, microRNA-1 (miR-1) and microRNA-21 (miR-21) have received some attention and have been associated with cardiac injury and cardioprotection. In this review article, we summarize the current knowledge of the function of these two miRs in the heart, their association with cardiac injury, and their potential cardioprotective roles and biomarker value. While this field has already been extensively studied, much remains to be done before research findings can be translated into clinical application for patient’s benefit.
Collapse
Affiliation(s)
- Branislav Kura
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (B.K.); (B.K.)
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 81372 Bratislava, Slovakia
| | - Barbora Kalocayova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (B.K.); (B.K.)
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg;
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (B.K.); (B.K.)
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 81372 Bratislava, Slovakia
- Correspondence: ; Tel.: +421-2-3229-5427
| |
Collapse
|
31
|
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: 0.8] [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
|
32
|
Step-by-Step Bioinformatics Analysis of Schistosoma mansoni Long Non-coding RNA Sequences. Methods Mol Biol 2020; 2151:109-133. [PMID: 32452000 DOI: 10.1007/978-1-0716-0635-3_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the last few years, long non-coding RNAs (lncRNAs) have been widely studied in humans, and their relevance for physiological and pathological conditions has been demonstrated. In parasites, there are only a few works, such as in Plasmodium falciparum, where it was shown that an lncRNA regulates the expression of a gene associated with immune system evasion, also indicating the relevance of understanding the role of this class of RNAs in parasites. In Schistosoma mansoni, in the last 2 years, there were four published articles related to the annotation of lncRNAs in different life cycle stages using RNA-Seq libraries. In order to make this process of lncRNA identification and annotation more accessible to biologists with no bioinformatics training, considering the growing number of S. mansoni RNA-Seq libraries publicly available from different sources, such as ovary tissues from bi-sex and single-sex infections, and the potential of lncRNAs as therapeutic targets, we provide this step-by-step protocol of lncRNA identification and quantification. This guide includes the download of RNA-Seq libraries from a public database and reads processing and mapping against the genome, transcript reconstruction, novel lncRNA identification, transcripts expression level determination, and the identification of differentially expressed lncRNAs.
Collapse
|
33
|
Jaquenod De Giusti C, Santalla M, Das S. Exosomal non-coding RNAs (Exo-ncRNAs) in cardiovascular health. J Mol Cell Cardiol 2019; 137:143-151. [PMID: 31669445 DOI: 10.1016/j.yjmcc.2019.09.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 09/05/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) play a role in the pathophysiological processes and in different diseases, including cardiovascular disease. Out of several categories of EVs, exosomes (smallest - 30 to 150 nm) are gaining most of the focus as the next generation of biomarkers and in therapeutic strategies. This is because exosomes can be differentiated from other types of EVs based on the expression of tetraspanin molecules on the surface. More importantly, exosomes can be traced back to the cell of origin by identifying the unique cellular marker(s) on the exosomal surface. Recently, several researchs have demonstrated an important and underappreciated mechanism of paracrine cell-cell communication involving exosomal transfer, and its subsequent functional impact on recipient cells. Exosomes are enriched in proteins, mRNAs, miRNAs, and other non-coding RNAs, which can potentially alter myocardial function. Additionally, different stages of tissue damage can also be identified by measuring these bioactive molecules in the circulation. There are several aspects of this new concept still unknown. Therefore, in this review, we have summarized the knowledge we have so far and highlighted the potential of this novel concept of next generation biomarkers and therapeutic intervention.
Collapse
Affiliation(s)
- Carolina Jaquenod De Giusti
- Centro de Investigaciones Cardiovasculares UNLP-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina.
| | - Manuela Santalla
- Centro de Investigaciones Cardiovasculares UNLP-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina; Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de Buenos Aires, Pergamino, Argentina
| | - Samarjit Das
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
34
|
Wu DM, Zhou ZK, Fan SH, Zheng ZH, Wen X, Han XR, Wang S, Wang YJ, Zhang ZF, Shan Q, Li MQ, Hu B, Lu J, Chen GQ, Hong XW, Zheng YL. Comprehensive RNA-Seq Data Analysis Identifies Key mRNAs and lncRNAs in Atrial Fibrillation. Front Genet 2019; 10:908. [PMID: 31632440 PMCID: PMC6783610 DOI: 10.3389/fgene.2019.00908] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 08/28/2019] [Indexed: 01/22/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are an emerging class of RNA species that may play a critical regulatory role in gene expression. However, the association between lncRNAs and atrial fibrillation (AF) is still not fully understood. In this study, we used RNA sequencing data to identify and quantify the both protein coding genes (PCGs) and lncRNAs. The high enrichment of these up-regulated genes in biological functions concerning response to virus and inflammatory response suggested that chronic viral infection may lead to activated inflammatory pathways, thereby alter the electrophysiology, structure, and autonomic remodeling of the atria. In contrast, the downregulated GO terms were related to the response to saccharides. To identify key lncRNAs involved in AF, we predicted lncRNAs regulating expression of the adjacent PCGs, and characterized biological function of the dysregulated lncRNAs. We found that two lncRNAs, ETF1P2, and AP001053.11, could interact with protein-coding genes (PCGs), which were implicated in AF. In conclusion, we identified key PCGs and lncRNAs, which may be implicated in AF, which not only improves our understanding of the roles of lncRNAs in AF, but also provides potentially functional lncRNAs for AF researchers.
Collapse
Affiliation(s)
- Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Zheng-Kun Zhou
- College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Zi-Hui Zheng
- State Key Laboratory Cultivation Base For TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Xin-Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Yong-Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Meng-Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Gui-Quan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Xiao-Wu Hong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| |
Collapse
|
35
|
Maciel LF, Morales-Vicente DA, Silveira GO, Ribeiro RO, Olberg GGO, Pires DS, Amaral MS, Verjovski-Almeida S. Weighted Gene Co-Expression Analyses Point to Long Non-Coding RNA Hub Genes at Different Schistosoma mansoni Life-Cycle Stages. Front Genet 2019; 10:823. [PMID: 31572441 PMCID: PMC6752179 DOI: 10.3389/fgene.2019.00823] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/09/2019] [Indexed: 01/21/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) (>200 nt) are expressed at levels lower than those of the protein-coding mRNAs, and in all eukaryotic model species where they have been characterized, they are transcribed from thousands of different genomic loci. In humans, some four dozen lncRNAs have been studied in detail, and they have been shown to play important roles in transcriptional regulation, acting in conjunction with transcription factors and epigenetic marks to modulate the tissue-type specific programs of transcriptional gene activation and repression. In Schistosoma mansoni, around 10,000 lncRNAs have been identified in previous works. However, the limited number of RNA-sequencing (RNA-seq) libraries that had been previously assessed, together with the use of old and incomplete versions of the S. mansoni genome and protein-coding transcriptome annotations, have hampered the identification of all lncRNAs expressed in the parasite. Here we have used 633 publicly available S. mansoni RNA-seq libraries from whole worms at different stages (n = 121), from isolated tissues (n = 24), from cell-populations (n = 81), and from single-cells (n = 407). We have assembled a set of 16,583 lncRNA transcripts originated from 10,024 genes, of which 11,022 are novel S. mansoni lncRNA transcripts, whereas the remaining 5,561 transcripts comprise 120 lncRNAs that are identical to and 5,441 lncRNAs that have gene overlap with S. mansoni lncRNAs already reported in previous works. Most importantly, our more stringent assembly and filtering pipeline has identified and removed a set of 4,293 lncRNA transcripts from previous publications that were in fact derived from partially processed mRNAs with intron retention. We have used weighted gene co-expression network analyses and identified 15 different gene co-expression modules. Each parasite life-cycle stage has at least one highly correlated gene co-expression module, and each module is comprised of hundreds to thousands lncRNAs and mRNAs having correlated co-expression patterns at different stages. Inspection of the top most highly connected genes within the modules’ networks has shown that different lncRNAs are hub genes at different life-cycle stages, being among the most promising candidate lncRNAs to be further explored for functional characterization.
Collapse
Affiliation(s)
- Lucas F Maciel
- Laboratório de Expressão Gênica em Eucariotos, Instituto Butantan, São Paulo, Brazil.,Programa Interunidades em Bioinformática, Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, Brazil
| | - David A Morales-Vicente
- Laboratório de Expressão Gênica em Eucariotos, Instituto Butantan, São Paulo, Brazil.,Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Gilbert O Silveira
- Laboratório de Expressão Gênica em Eucariotos, Instituto Butantan, São Paulo, Brazil.,Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Raphael O Ribeiro
- Laboratório de Expressão Gênica em Eucariotos, Instituto Butantan, São Paulo, Brazil.,Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Giovanna G O Olberg
- Laboratório de Expressão Gênica em Eucariotos, Instituto Butantan, São Paulo, Brazil
| | - David S Pires
- Laboratório de Expressão Gênica em Eucariotos, Instituto Butantan, São Paulo, Brazil
| | - Murilo S Amaral
- Laboratório de Expressão Gênica em Eucariotos, Instituto Butantan, São Paulo, Brazil
| | - Sergio Verjovski-Almeida
- Laboratório de Expressão Gênica em Eucariotos, Instituto Butantan, São Paulo, Brazil.,Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
Affiliation(s)
- Amela Jusic
- From the Department of Biology, Faculty of Natural Sciences and Mathematics, University of Tuzla, Bosnia and Herzegovina (A.J.)
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health (Y.D.)
| | | |
Collapse
|
37
|
Long Noncoding Competing Endogenous RNA Networks in Age-Associated Cardiovascular Diseases. Int J Mol Sci 2019; 20:ijms20123079. [PMID: 31238513 PMCID: PMC6627372 DOI: 10.3390/ijms20123079] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/17/2019] [Accepted: 06/21/2019] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the most serious health problem in the world, displaying high rates of morbidity and mortality. One of the main risk factors for CVDs is age. Indeed, several mechanisms are at play during aging, determining the functional decline of the cardiovascular system. Aging cells and tissues are characterized by diminished autophagy, causing the accumulation of damaged proteins and mitochondria, as well as by increased levels of oxidative stress, apoptosis, senescence and inflammation. These processes can induce a rapid deterioration of cellular quality-control systems. However, the molecular mechanisms of age-associated CVDs are only partially known, hampering the development of novel therapeutic strategies. Evidence has emerged indicating that noncoding RNAs (ncRNAs), such as long ncRNAs (lncRNAs) and micro RNAs (miRNAs), are implicated in most patho-physiological mechanisms. Specifically, lncRNAs can bind miRNAs and act as competing endogenous-RNAs (ceRNAs), therefore modulating the levels of the mRNAs targeted by the sponged miRNA. These complex lncRNA/miRNA/mRNA networks, by regulating autophagy, apoptosis, necrosis, senescence and inflammation, play a crucial role in the development of age-dependent CVDs. In this review, the emerging knowledge on lncRNA/miRNA/mRNA networks will be summarized and the way in which they influence age-related CVDs development will be discussed.
Collapse
|
38
|
Liu X, Ma Y, Yin K, Li W, Chen W, Zhang Y, Zhu C, Li T, Han B, Liu X, Wang S, Zhou Z. Long non-coding and coding RNA profiling using strand-specific RNA-seq in human hypertrophic cardiomyopathy. Sci Data 2019; 6:90. [PMID: 31197155 PMCID: PMC6565738 DOI: 10.1038/s41597-019-0094-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/15/2019] [Indexed: 01/09/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) represents one of the most common heritable heart diseases. However, the signalling pathways and regulatory networks underlying the pathogenesis of HCM remain largely unknown. Here, we present a strand-specific RNA-seq dataset for both coding and lncRNA profiling in myocardial tissues from 28 HCM patients and 9 healthy donors. This dataset constitutes a valuable resource for the community to examine the dysregulated coding and lncRNA genes in HCM versus normal conditions.
Collapse
Affiliation(s)
- Xuanyu Liu
- Center of Laboratory Medicine, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yi Ma
- Center of Laboratory Medicine, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Kunlun Yin
- Center of Laboratory Medicine, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Wenke Li
- Center of Laboratory Medicine, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Wen Chen
- Center of Laboratory Medicine, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yujing Zhang
- Center of Laboratory Medicine, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Changsheng Zhu
- Departments of Cardiovascular Surgery, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Tianjiao Li
- Center of Laboratory Medicine, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Bianmei Han
- Center of Laboratory Medicine, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xuewen Liu
- Center of Laboratory Medicine, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Shuiyun Wang
- Departments of Cardiovascular Surgery, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Zhou Zhou
- Center of Laboratory Medicine, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
39
|
Li Y, Zhang D, Zhang Y, Xu X, Bi L, Zhang M, Yu B, Zhang Y. Association of lncRNA polymorphisms with triglyceride and total cholesterol levels among myocardial infarction patients in Chinese population. Gene 2019; 724:143684. [PMID: 30898706 DOI: 10.1016/j.gene.2019.02.085] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 01/21/2019] [Accepted: 02/22/2019] [Indexed: 01/02/2023]
Abstract
AIM The long noncoding RNAs (lncRNAs) have gradually been reported to be an important class of RNAs with pivotal roles in the development and progression of myocardial infarction (MI). In this study, we hypothesized that genetic variant of cyclin-dependent kinase inhibitor 2B antisense RNA (ANRIL) and metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) may affect the prognosis of MI patients. METHODS The study included 401 Han Chinese MI patients and 409 controls. Four lncRNA tag single nucleotide polymorphisms (SNPs)-ANRIL rs9632884 and rs1537373, MALAT1 rs619586 and rs3200401-were selected. SNP genotyping was performed by an improved multiplex ligation detection reaction assay. RESULTS rs9632884 and rs3200401 SNPs were significantly associated with lipid levels in both controls and MI patients (P < 0.003-0.046). Several SNPs interacted with sex and age to modify total cholesterol, low-density lipoprotein cholesterol, and creatinine levels to modify the risk of MI. No association between the lncRNAs SNPs and susceptibility to MI was found (P > 0.05 for all). CONCLUSIONS Taken together, this study provides additional evidence that genetic variation of the ANRIL rs9632884 and MALAT1 rs3200401 can mediate lipid levels in MI patients.
Collapse
Affiliation(s)
- Yilan Li
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, China
| | - Dandan Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, China
| | - Yanxiu Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xueming Xu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Lei Bi
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Meiling Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, China
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, China
| | - Yao Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
40
|
Cardiovascular inflammation: RNA takes the lead. J Mol Cell Cardiol 2019; 129:247-256. [PMID: 30880251 DOI: 10.1016/j.yjmcc.2019.03.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/19/2022]
Abstract
Inflammation has recently gained tremendous attention as a key contributor in several chronic diseases. While physiological inflammation is essential to counter a wide variety of damaging stimuli and to improve wound healing, dysregulated inflammation such as in the myocardium and vasculature can promote cardiovascular diseases. Given the high severity, prevalence, and economic burden of these diseases, understanding the factors involved in the regulation of physiological inflammation is essential. Like other complex biological phenomena, RNA-based processes are emerging as major regulators of inflammatory responses. Among such processes are cis-regulatory elements in the mRNA of inflammatory genes, noncoding RNAs directing the production or localization of inflammatory cytokines/chemokines, or pathogenic RNA driving inflammatory responses. In this review, we describe several specific RNA-based molecular mechanisms by which physiological inflammation pertaining to cardiovascular diseases is regulated. These include the role of AU-rich element-containing mRNAs, long non-coding RNAs, microRNAs, and viral RNAs.
Collapse
|
41
|
Zhuang Y, Li T, Zhuang Y, Li Z, Yang W, Huang Q, Li D, Wu H, Zhang G, Yang T, Zhan L, Pan Z, Lu Y. Involvement of lncR-30245 in Myocardial Infarction-Induced Cardiac Fibrosis Through Peroxisome Proliferator-Activated Receptor-γ-Mediated Connective Tissue Growth Factor Signalling Pathway. Can J Cardiol 2019; 35:480-489. [PMID: 30935639 DOI: 10.1016/j.cjca.2019.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 02/10/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are emerging as important mediators of cardiac pathophysiology. The aim of the present study is to investigate the effects of lncR-30245, an lncRNA, on cardiac fibrogenesis and the underlying mechanism. METHODS Myocardial infarction (MI) and transforming growth factor (TGF)-β1 were used to induce fibrotic phenotypes. Cardiac fibrosis was detected by Masson's trichrome staining. Cardiac function was evaluated by echocardiography. Western blot, quantitative reverse transcription-polymerase chain reaction, and pharmacological approaches were used to investigate the role of lncR-30245 in cardiac fibrogenesis. RESULTS Expression of lncR-30245 was significantly increased in MI hearts and TGF-β1-treated cardiac fibroblasts (CFs). LncR-30245 was mainly located in the cytoplasm. Overexpression of lncR-30245 promoted collagen production and CF proliferation. Knockdown of lncR-30245 significantly inhibited TGF-β1-induced collagen production and CF proliferation. LncR-30245 overexpression inhibited the antifibrotic role of peroxisome proliferator-activated receptor (PPAR)-γ and increased connective tissue growth factor (CTGF) expression, whereas lncR-30245 knockdown exerted the opposite effects. Rosiglitazone, a PPAR-γ agonist, significantly inhibited lncR-30245-induced CTGF upregulation and collagen production in CFs. In contrast, T0070907, a PPAR-γ antagonist, attenuated the inhibitory effects of lncR-30245 small interfering RNA (siRNA) on TGF-β1-induced CTGF expression and collagen production. LncR-30245 knockdown significantly enhanced ejection fraction and fractional shortening and attenuated cardiac fibrosis in MI mice. CONCLUSION Our study indicates that the lncR-30245/PPAR-γ/CTGF pathway mediates MI-induced cardiac fibrosis and might be a therapeutic target for various cardiac diseases associated with fibrosis.
Collapse
Affiliation(s)
- Yuting Zhuang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Tingting Li
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Yanan Zhuang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Zhuoyun Li
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Wanqi Yang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Qihe Huang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Danyang Li
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Hao Wu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Guiye Zhang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Ti Yang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Linfeng Zhan
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Zhenwei Pan
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China.
| | - Yanjie Lu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, P. R. China.
| |
Collapse
|
42
|
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: 1.5] [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
|
43
|
Non-Coding RNAs to Aid in Neurological Prognosis after Cardiac Arrest. Noncoding RNA 2018; 4:ncrna4040042. [PMID: 30567385 PMCID: PMC6316658 DOI: 10.3390/ncrna4040042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/07/2018] [Accepted: 12/12/2018] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease in general, and sudden cardiac death in particular, have an enormous socio-economic burden worldwide. Despite significant efforts to improve cardiopulmonary resuscitation, survival rates remain low. Moreover, patients who survive to hospital discharge have a high risk of developing severe physical or neurological symptoms. Being able to predict outcomes after resuscitation from cardiac arrest would make it possible to tailor healthcare approaches, thereby maximising efforts for those who would mostly benefit from aggressive therapy. However, the identification of patients at risk of poor recovery after cardiac arrest is still a challenging task which could be facilitated by novel biomarkers. Recent investigations have recognised the potential of non-coding RNAs to aid in outcome prediction after cardiac arrest. In this review, we summarize recent discoveries and propose a handful of novel perspectives for the use of non-coding RNAs to predict outcome after cardiac arrest, discussing their use for precision medicine.
Collapse
|
44
|
The emerging role of lncRNAs in inflammatory bowel disease. Exp Mol Med 2018; 50:1-14. [PMID: 30523244 PMCID: PMC6283835 DOI: 10.1038/s12276-018-0188-9] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/28/2018] [Accepted: 09/11/2018] [Indexed: 12/19/2022] Open
Abstract
Dysregulation of long noncoding RNA (lncRNA) expression is linked to the development of various diseases. Recently, an emerging body of evidence has indicated that lncRNAs play important roles in the pathogenesis of inflammatory bowel diseases (IBDs), including Crohn’s disease (CD) and ulcerative Colitis (UC). In IBD, lncRNAs have been shown to be involved in diverse processes, including the regulation of intestinal epithelial cell apoptosis, association with lipid metabolism, and cell–cell interactions, thereby enhancing inflammation and the functional regulation of regulatory T cells. In this review, we aim to summarize the current knowledge regarding the role of lncRNAs in IBD and highlight potential avenues for future investigation. We also collate potentially immune-relevant, IBD-associated lncRNAs identified through a built-by association analysis with respect to their neighboring protein-coding genes within IBD-susceptible loci. We further underscore their importance by highlighting their enrichment for various aspects of immune system regulation, including antigen processing/presentation, immune cell proliferation and differentiation, and chronic inflammatory responses. Finally, we summarize the potential of lncRNAs as diagnostic biomarkers in IBD. Studying long noncoding RNAs (lncRNAs) may improve diagnosis and treatment of inflammatory bowel disease (IBD). These RNAs are found between genes in DNA regions previously thought to be “junk,” and have recently been shown to be important in development of various diseases. IBD, which includes both Crohn’s disease and ulcerative colitis, damages the digestive tract lining, causing pain and chronic diarrhea. A better understanding of IBD’s complex causes is needed to identify more effective treatments. Flemming Pociot at the Steno Diabetes Center in Gentofte, Denmark, and co-workers reviewed recent research linking lncRNAs and IBD. They discuss how lncRNAs’ roles in immunity and inflammation influence IBD development, describing how particular lncRNAs are related to IBD. Promising avenues for further research are highlighted, including the use of lncRNAs as biomarkers of IBD, which can be difficult to diagnose.
Collapse
|
45
|
Bacci L, Barbati SA, Colussi C, Aiello A, Isidori AM, Grassi C, Pontecorvi A, Farsetti A, Gaetano C, Nanni S. Sildenafil normalizes MALAT1 level in diabetic cardiomyopathy. Endocrine 2018; 62:259-262. [PMID: 29691810 DOI: 10.1007/s12020-018-1599-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 04/09/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Lorenza Bacci
- Institute of Medical Pathology, Università Cattolica di Roma, 00168, Rome, Italy
| | - Saviana A Barbati
- Institute of Human Physiology, Università Cattolica di Roma, 00168, Rome, Italy
| | - Claudia Colussi
- Institute of Cell Biology and Neurobiology, National Research Council, 00143, Rome, Italy
| | - Aurora Aiello
- Institute of Medical Pathology, Università Cattolica di Roma, 00168, Rome, Italy
- Institute of Cell Biology and Neurobiology, National Research Council, 00143, Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, "Sapienza" University, 00161, Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica di Roma, 00168, Rome, Italy
- Fondazione Policlinico Universitario Gemelli, 00168, Rome, Italy
| | - Alfredo Pontecorvi
- Institute of Medical Pathology, Università Cattolica di Roma, 00168, Rome, Italy
- Fondazione Policlinico Universitario Gemelli, 00168, Rome, Italy
| | - Antonella Farsetti
- Institute of Cell Biology and Neurobiology, National Research Council, 00143, Rome, Italy
| | - Carlo Gaetano
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri, via Maugeri 4, 27100, Pavia, Italy.
| | - Simona Nanni
- Institute of Medical Pathology, Università Cattolica di Roma, 00168, Rome, Italy.
| |
Collapse
|
46
|
Hermans-Beijnsberger S, van Bilsen M, Schroen B. Long non-coding RNAs in the failing heart and vasculature. Noncoding RNA Res 2018; 3:118-130. [PMID: 30175285 PMCID: PMC6114261 DOI: 10.1016/j.ncrna.2018.04.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023] Open
Abstract
Following completion of the human genome, it became evident that the majority of our DNA is transcribed into non-coding RNAs (ncRNAs) instead of protein-coding messenger RNA. Deciphering the function of these ncRNAs, including both small- and long ncRNAs (lncRNAs), is an emerging field of research. LncRNAs have been associated with many disorders and a number have been identified as key regulators in the development and progression of disease, including cardiovascular disease (CVD). CVD causes millions of deaths worldwide, annually. Risk factors include coronary artery disease, high blood pressure and ageing. In this review, we will focus on the roles of lncRNAs in the cellular and molecular processes that underlie the development of CVD: cardiomyocyte hypertrophy, fibrosis, inflammation, vascular disease and ageing. Finally, we discuss the biomarker and therapeutic potential of lncRNAs.
Collapse
Affiliation(s)
- Steffie Hermans-Beijnsberger
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - Marc van Bilsen
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
- Department of Physiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - Blanche Schroen
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
47
|
Zhu P, Yang M, Ren H, Shen G, Chen J, Zhang J, Liu J, Sun C. Long noncoding RNA MALAT1 downregulates cardiac transient outward potassium current by regulating miR-200c/HMGB1 pathway. J Cell Biochem 2018; 119:10239-10249. [PMID: 30145795 DOI: 10.1002/jcb.27366] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
The dysregulation of long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) participates in the remodeling of electrophysiological/ion channel in cardiomyocytes during arrhythmia. The lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is reported to be highly expressed in myocardial ischemia-reperfusion injury and offsets cardioprotective effects of fentanyl. However, the roles of MALAT1 and its related miRNAs during arrhythmia are poorly understood. In this study, the overexpression of MALAT1 was firstly indicated in cardiomyocytes from arrhythmic model rats. After downregulation of MALAT1 by RNA interference, transient outward potassium current (Ito), peak current density, and the levels of Kv4.2 and Kv4.3 channel proteins were increased in rat cardiomyocytes. Then, miR-200c was predicted and convinced to be a direct target of MALAT1, and high-mobility group box 1 (HMGB1) was verified to be a target of miR-200c during arrhythmia. HMGB1 expression reduced by the knockdown of MALAT1 was further decreased by miR-200c overexpression. In addition, cardiac Ito, peak current density, and the levels of Kv4.2 and Kv4.3 in arrhythmic model rats were detected to be negatively correlated with the expression of HMGB1, and to be positively with miR-200c expression. Taken together, these results suggested that MALAT1 may act as a competing endogenous RNA for miR-200c to upregulate the expression of HMGB1 and downregulate cardiac Ito.
Collapse
Affiliation(s)
- Peng Zhu
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Medical College of Xi'an Jiaotong University, Xi'an, China.,Department of Cardiovascular Medicine, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China
| | - Manli Yang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China
| | - Hui Ren
- Department of Cardiovascular Medicine, Ankang Central Hospital, Ankang, China
| | - Guidong Shen
- Department of Cardiovascular Medicine, Ankang Central Hospital, Ankang, China
| | - Jinye Chen
- Department of Cardiovascular Medicine, Ankang Central Hospital, Ankang, China
| | - Junkang Zhang
- Department of Cardiovascular Medicine, Ankang Central Hospital, Ankang, China
| | - Jun Liu
- Department of Pathology, Ankang Central Hospital, Ankang, China
| | - Chaofeng Sun
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Medical College of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
48
|
Zhuang Y, Li T, Zhuang Y, Li Z, Yang W, Huang Q, Li D, Wu H, Zhang G, Yang T, Zhan L, Pan Z, Lu Y. WITHDRAWN: Suppression of lncR-30245 alleviates myocardial infarction induced cardiac fibrosis via the PPAR-γ/CTGF pathway. Can J Cardiol 2018. [DOI: 10.1016/j.cjca.2018.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
49
|
Gomes CP, Salgado-Somoza A, Creemers EE, Dieterich C, Lustrek M, Devaux Y. Circular RNAs in the cardiovascular system. Noncoding RNA Res 2018; 3:1-11. [PMID: 30159434 PMCID: PMC6084836 DOI: 10.1016/j.ncrna.2018.02.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/16/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023] Open
Abstract
Until recently considered as rare, circular RNAs (circRNAs) are emerging as important regulators of gene expression. They are ubiquitously expressed and represent a novel branch of the family of non-coding RNAs. Recent investigations showed that circRNAs are regulated in the cardiovascular system and participate in its physiological and pathological development. In this review article, we will provide an overview of the role of circRNAs in cardiovascular health and disease. After a description of the biogenesis of circRNAs, we will summarize what is known of the expression, regulation and function of circRNAs in the cardiovascular system. We will then address some technical aspects of circRNAs research, discussing how artificial intelligence may aid in circRNAs research. Finally, the potential of circRNAs as biomarkers of cardiovascular disease will be addressed and directions for future research will be proposed.
Collapse
Key Words
- Artificial intelligence
- Biomarker
- CRISPR, clustered regularly interspaced short palindromic repeats
- CV, cardiovascular
- Cardiovascular disease
- Cardiovascular system
- Circular RNAs
- DCM, dilated cardiomyopathy
- EMT, epithelial-mesenchymal transition
- Non-coding RNAs
- RNA-seq, RNA sequencing
- RPAD, RNase R treatment followed by polyadenylation and poly(A)+ RNA depletion
- RT-qPCR, reverse transcription quantitative polymerase chain reaction
- circRNAs, circular RNAs
- lncRNAs, long non-coding RNAs
- miRNAs, microRNAs
- ncRNAs, non-coding RNAs
Collapse
Affiliation(s)
- Clarissa P.C. Gomes
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | | | - Esther E. Creemers
- Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Christoph Dieterich
- German Center for Cardiovascular Research, University Hospital Heidelberg, Heidelberg, Germany
| | - Mitja Lustrek
- Department of Intelligent Systems, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | | |
Collapse
|
50
|
Microarray analysis of lung long non-coding RNAs in cigarette smoke-exposed mouse model. Oncotarget 2017; 8:115647-115656. [PMID: 29383188 PMCID: PMC5777800 DOI: 10.18632/oncotarget.23362] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/05/2017] [Indexed: 02/05/2023] Open
Abstract
Several studies have demonstrated the function of long non‑coding RNAs (lncRNAs) in various biological processes, yet their role underlying the susceptibility to cigarette smoke (CS)-induced airway inflammation remains limited. In the present study, we aimed to profile the expression of lncRNAs and mRNAs in CS-exposed mice. C57BL/6 mice were assigned into a single cigarette-smoking machine with or without CS exposure for 4 weeks, followed by lung tissue harvest and RNA isolation. Microarray analysis identified 108 lncRNAs and 119 mRNAs with differential expression levels in CS-exposed mouse lung tissue compared with those in control mice. The expression patterns of several lncRNAs were further confirmed by qRT-PCR. GO and pathway analyses showed that the altered mRNAs were mainly related to the processes of immune response, defense response and cell chemotaxis, cytokine-cytokine receptor interaction and chemokine signaling pathway. Moreover, a single lncRNA may co-expressed with several mRNAs, and so was the mRNA. Our findings uncovered the expression profile of lncRNAs and mRNAs in the lungs of CS-exposed mice, which may offer new insights into pathogenesis of CS-associated airway inflammatory disorders.
Collapse
|