1
|
He YZG, Wang YX, Ma JS, Li RN, Wang J, Lian TY, Zhou YP, Yang HP, Sun K, Jing ZC. MicroRNAs and their regulators: Potential therapeutic targets in pulmonary arterial hypertension. Vascul Pharmacol 2023; 153:107216. [PMID: 37699495 DOI: 10.1016/j.vph.2023.107216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 08/26/2023] [Accepted: 09/03/2023] [Indexed: 09/14/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a complex and progressive disease characterized by pulmonary arterial remodeling. Despite that current combination therapy has shown improvement in morbidity and mortality, a better deciphering of the underlying pathological mechanisms and novel therapeutic targets is urgently needed to combat PAH. MicroRNA, the critical element in post-transcription mechanisms, mediates cellular functions mainly by tuning downstream target gene expression. Meanwhile, upstream regulators can regulate miRNAs in synthesis, transcription, and function. In vivo and in vitro studies have suggested that miRNAs and their regulators are involved in PAH. However, the miRNA-related regulatory mechanisms governing pulmonary vascular remodeling and right ventricular dysfunction remain elusive. Hence, this review summarized the controversial roles of miRNAs in PAH pathogenesis, focused on different miRNA-upstream regulators, including transcription factors, regulatory networks, and environmental stimuli, and finally proposed the prospects and challenges for the therapeutic application of miRNAs and their regulators in PAH treatment.
Collapse
Affiliation(s)
- Yang-Zhi-Ge He
- Center for bioinformatics, National Infrastructures for Translational Medicine, Institute of Clinical Medicine & Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Yi-Xuan Wang
- Laboratory Department of Qingzhou People's Hospital, Qingzhou 262500, Shandong, China
| | - Jing-Si Ma
- Department of School of Pharmacy, Henan University, Kaifeng 475100, Henan, China
| | - Ruo-Nan Li
- Department of School of Pharmacy, Henan University, Kaifeng 475100, Henan, China
| | - Jia Wang
- Department of Medical Laboratory, Weifang Medical University, Weifang 261053, Shandong, China
| | - Tian-Yu Lian
- Medical Science Research Center, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Yu-Ping Zhou
- Department of Cardiology, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Beijing 100730, China
| | - Hao-Pu Yang
- Tsinghua University School of Medicine, Beijing 100084, China
| | - Kai Sun
- Medical Science Research Center, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China.
| | - Zhi-Cheng Jing
- Department of Cardiology, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Beijing 100730, China.
| |
Collapse
|
2
|
Wang D, Luo MY, Tian Y, Zhang J, Liang N, Li NP, Gong SX, Wang AP. Critical miRNAs in regulating pulmonary hypertension: A focus on Signaling pathways and therapeutic Targets. Anal Biochem 2023:115228. [PMID: 37393975 DOI: 10.1016/j.ab.2023.115228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/19/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
Pulmonary hypertension (PH) is complex disease as a result of obstructive pulmonary arterial remodeling, which in turn results in elevated pulmonary arterial pressure (PAP) and subsequent right ventricular heart failure, eventually leading to premature death. However, there is still a lack of a diagnostic blood-based biomarker and therapeutic target for PH. Because of the difficulty of diagnosis, new and more easily accessible prevention and treatment strategy are being explored. New target and diagnosis biomarkers should also allow for early diagnosis. In biology, miRNAs are short endogenous RNA molecules that are not coding. It is known that miRNAs can regulate gene expression and affect a variety of biological processes. Besides, miRNAs have been proven to be a crucial factor in PH pathogenesis. miRNAs have various effects on pulmonary vascular remodeling and are expressed differentially in various pulmonary vascular cells. Nowadays, it has been shown to be critical in the functions of different miRNAs in the pathogenesis of PH. Therefore, clarifying the mechanism of miRNAs regulating pulmonary vascular remodeling is of great importance to explore new therapeutic targets of PH and improve the survival qualify and time of patients. This review is focused on the role, mechanism, and potential therapeutic targets of miRNAs in PH and puts forward possible clinical treatment strategies.
Collapse
Affiliation(s)
- Di Wang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China
| | - Meng-Yi Luo
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China; Institute of Clinical Research, Department of Clinical Laboratory, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, PR China
| | - Ying Tian
- Institute of Clinical Research, Department of Clinical Laboratory, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, PR China
| | - Jing Zhang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China
| | - Na Liang
- Department of Anesthesiology, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, PR China
| | - Nan-Ping Li
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China; Department of Anesthesiology, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, PR China
| | - Shao-Xin Gong
- Department of Pathology, First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China.
| | - Ai-Ping Wang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China; Institute of Clinical Research, Department of Clinical Laboratory, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, PR China.
| |
Collapse
|
3
|
Bai X, Zhang H, Li Z, Chen O, He H, Jia X, Zou L. Platelet-derived extracellular vesicles encapsulate microRNA-34c-5p to ameliorate inflammatory response of coronary artery endothelial cells via PODXL-mediated P38 MAPK signaling pathway. Nutr Metab Cardiovasc Dis 2022; 32:2424-2438. [PMID: 36096977 DOI: 10.1016/j.numecd.2022.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/30/2022] [Accepted: 06/14/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND AND AIMS Low-grade chronic inflammation was reported to serve as a distinctive pathophysiologic feature of coronary artery disease (CAD), the leading cause of death around the world. Herein, the current study aimed to explore whether and how microRNA-34c-5p (miR-34c-5p), a miRNA enriched in extracellular vesicles (EVs) originated from the activated platelet (PLT-EVs), affects the inflammation of human coronary artery endothelial cells (HCAECs). METHODS AND RESULTS HCAECs were established as an in vitro cell model using oxidized low-density lipoprotein (ox-LDL). miR-34c-5p, an abundant miRNA in PLT-EVs, can be transferred to HCAECs and target PODXL by binding to its 3'UTR. Gain- and loss-of-function experiments of miR-34c-5p and podocalyxin (PODXL) were performed in ox-LDL-induced HCAECs. Subsequently, HCAECs were subjected to co-culture with PLT-EVs, followed by detection of the expression patterns of key pro-inflammatory factors. Either miR-34c-5p mimic or PLT-EVs harboring miR-34c-5p attenuated the ox-LDL-evoked inflammation in HCAECs by suppressing interleukin-1β (IL-1β), IL-6 and tumor necrosis factor-α (TNF-α). By blocking the P38 MAPK signaling pathway, miR-34c-5p-mediated depletion of PODXL contributed to protection against ox-LDL-induced inflammation. In vitro findings were further validated by findings observed in ApoE knock-out mice. Additionally, miR-34c-5p in PLT-EVs showed an athero-protective role in the murine model. CONCLUSION Altogether, our findings highlighted that miR-34c-5p in PLT-EVs could alleviate inflammation response in HCAECs by targeting PODXL and inactivation of the P38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Xuetao Bai
- Department of Anaesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, PR China
| | - Hao Zhang
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China
| | - Zhiguo Li
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China
| | - Ou Chen
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China
| | - Hengpeng He
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China
| | - Xiukun Jia
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China
| | - Lijuan Zou
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China.
| |
Collapse
|
4
|
Qian J, Yu J, Zhu X, Liang S. MiR-335 promotes corneal neovascularization by Targeting EGFR. BMC Ophthalmol 2022; 22:267. [PMID: 35701740 PMCID: PMC9199176 DOI: 10.1186/s12886-022-02481-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022] Open
Abstract
Background Corneal neovascularization (CRNV) is a severe threat to the vision of people. MicroRNA-335 (miR-335) has the function of facilitating angiogenesis. However, whether miR-335 regulates the progression of CRNV remains unclear. Methods The miR-335 expressions in CRNV rats induced by corneal suture and HUVECs induced by b-FGF were detected by quantitative real-time PCR. For the miR-335 function, wound healing and tube formation assays were performed. For the miR-335 mechanism, a dual-luciferase reporter gene assay was conducted. Besides, for the epidermal growth factor receptor (EGFR) function, Cell Counting Kit-8 and wound healing assays were performed. Meanwhile, the rescue assay was used to assess the miR-335/EGFR function in the migration and angiogenesis of b-FGF-treated HUVECs. Results Functionally, the miR-335 knockdown weakened the migration and angiogenesis of b-FGF-treated HUVECs, while the miR-335 overexpression showed an opposite trend. Mechanistically, miR-335 interacted with EGFR and negatively regulated the expression of EGFR. The rescue assay illustrated that miR-335 regulated the migration and angiogenesis of b-FGF-treated HUVECs through EGFR. Conclusions In general, our data confirmed that miR-335 facilitated the process of CRNV by targeting EGFR. Supplementary Information The online version contains supplementary material available at 10.1186/s12886-022-02481-0.
Collapse
Affiliation(s)
- Jingjing Qian
- Department of Ophthalmology, Affiliated Hospital of Nantong University, No. 20 Xisi Road, Nantong, 226001, Jiangsu Province, China
| | - Junbo Yu
- Department of Trauma Center, Affiliated Hospital of Nantong University, No. 20 Xisi Road, Nantong, 226001, Jiangsu Province, China
| | - Xi Zhu
- Department of Ophthalmology, Affiliated Hospital of Nantong University, No. 20 Xisi Road, Nantong, 226001, Jiangsu Province, China
| | - Shu Liang
- Department of Ophthalmology, Affiliated Hospital of Nantong University, No. 20 Xisi Road, Nantong, 226001, Jiangsu Province, China.
| |
Collapse
|
5
|
Wiedmann F, Kraft M, Kallenberger S, Büscher A, Paasche A, Blochberger PL, Seeger T, Jávorszky N, Warnecke G, Arif R, Kremer J, Karck M, Frey N, Schmidt C. MicroRNAs Regulate TASK-1 and Are Linked to Myocardial Dilatation in Atrial Fibrillation. J Am Heart Assoc 2022; 11:e023472. [PMID: 35301863 PMCID: PMC9075420 DOI: 10.1161/jaha.121.023472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia. However, underlying molecular mechanisms are insufficiently understood. Previous studies suggested that microRNA (miRNA) dependent gene regulation plays an important role in the initiation and maintenance of AF. The 2‐pore‐domain potassium channel TASK‐1 (tandem of P domains in a weak inward rectifying K+ channel–related acid sensitive K+ channel 1) is an atrial‐specific ion channel that is upregulated in AF. Inhibition of TASK‐1 current prolongs the atrial action potential duration to similar levels as in patients with sinus rhythm. Here, we hypothesize that miRNAs might be responsible for the regulation of KCNK3 that encodes for TASK‐1. Methods and Results We selected miRNAs potentially regulating KCNK3 and studied their expression in atrial tissue samples obtained from patients with sinus rhythm, paroxysmal AF, or permanent/chronic AF. MiRNAs differentially expressed in AF were further investigated for their ability to regulate KCNK3 mRNA and TASK‐1 protein expression in human induced pluripotent stem cells, transfected with miRNA mimics or inhibitors. Thereby, we observed that miR‐34a increases TASK‐1 expression and current and further decreases the resting membrane potential of Xenopus laevis oocytes, heterologously expressing hTASK‐1. Finally, we investigated associations between miRNA expression in atrial tissues and clinical parameters of our patient cohort. A cluster containing AF stage, left ventricular end‐diastolic diameter, left ventricular end‐systolic diameter, left atrial diameter, atrial COL1A2 (collagen alpha‐2(I) chain), and TASK‐1 protein level was associated with increased expression of miR‐25, miR‐21, miR‐34a, miR‐23a, miR‐124, miR‐1, and miR‐29b as well as decreased expression of miR‐9 and miR‐485. Conclusions These results suggest an important pathophysiological involvement of miRNAs in the regulation of atrial expression of the TASK‐1 potassium channel in patients with atrial cardiomyopathy.
Collapse
Affiliation(s)
- Felix Wiedmann
- Department of Cardiology Heidelberg University Hospital Heidelberg Germany.,DZHK (German Center for Cardiovascular Research) Partner Site Heidelberg/Mannheim University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders Heidelberg University Hospital Heidelberg Germany
| | - Manuel Kraft
- Department of Cardiology Heidelberg University Hospital Heidelberg Germany.,DZHK (German Center for Cardiovascular Research) Partner Site Heidelberg/Mannheim University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders Heidelberg University Hospital Heidelberg Germany
| | - Stefan Kallenberger
- Digital Health Center Berlin Institute of Health (BIH) and Charité Berlin Germany.,Department of Medical Oncology National Center for Tumor DiseasesHeidelberg University Hospital Heidelberg Germany.,Health Data Science UnitMedical Faculty Heidelberg Heidelberg Germany
| | - Antonius Büscher
- Department for Cardiology II: Electrophysiology University Hospital Münster Münster Germany
| | - Amelie Paasche
- Department of Cardiology Heidelberg University Hospital Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders Heidelberg University Hospital Heidelberg Germany
| | - Pablo L Blochberger
- Department of Cardiology Heidelberg University Hospital Heidelberg Germany.,DZHK (German Center for Cardiovascular Research) Partner Site Heidelberg/Mannheim University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders Heidelberg University Hospital Heidelberg Germany
| | - Timon Seeger
- Department of Cardiology Heidelberg University Hospital Heidelberg Germany
| | - Natasa Jávorszky
- Department of Cardiology Heidelberg University Hospital Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders Heidelberg University Hospital Heidelberg Germany
| | - Gregor Warnecke
- Department of Cardiac Surgery University of Heidelberg Germany
| | - Rawa Arif
- Department of Cardiac Surgery University of Heidelberg Germany
| | - Jamila Kremer
- Department of Cardiac Surgery University of Heidelberg Germany
| | - Matthias Karck
- Department of Cardiac Surgery University of Heidelberg Germany
| | - Norbert Frey
- Department of Cardiology Heidelberg University Hospital Heidelberg Germany.,DZHK (German Center for Cardiovascular Research) Partner Site Heidelberg/Mannheim University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders Heidelberg University Hospital Heidelberg Germany
| | - Constanze Schmidt
- Department of Cardiology Heidelberg University Hospital Heidelberg Germany.,DZHK (German Center for Cardiovascular Research) Partner Site Heidelberg/Mannheim University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders Heidelberg University Hospital Heidelberg Germany
| |
Collapse
|
6
|
Hua CC, Liu XM, Liang LR, Wang LF, Zhong JC. Targeting the microRNA-34a as a Novel Therapeutic Strategy for Cardiovascular Diseases. Front Cardiovasc Med 2022; 8:784044. [PMID: 35155600 PMCID: PMC8828972 DOI: 10.3389/fcvm.2021.784044] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are still the main cause of morbidity and mortality worldwide and include a group of disorders varying from vasculature, myocardium, arrhythmias and cardiac development. MicroRNAs (miRs) are endogenous non-coding RNAs with 18–23 nucleotides that regulate gene expression. The miR-34 family, including miR-34a/b/c, plays a vital role in the regulation of myocardial physiology and pathophysiological processes. Recently, miR-34a has been implicated in cardiovascular fibrosis, dysfunction and related cardiovascular disorders as an essential regulator. Interestingly, there is a pivotal link among miR-34a, cardiovascular fibrosis, and Smad4/TGF-β1 signaling. Notably, both loss-of-function and gain-of-function approaches identified the critical roles of miR-34a in cardiovascular apoptosis, autophagy, inflammation, senescence and remodeling by modulating multifunctional signaling pathways. In this article, we focus on the current understanding of miR-34a in biogenesis, its biological effects and its implications for cardiac pathologies including myocardial infarction, heart failure, ischaemia reperfusion injury, cardiomyopathy, atherosclerosis, hypertension and atrial fibrillation. Thus, further understanding of the effects of miR-34a on cardiovascular diseases will aid the development of effective interventions. Targeting for miR-34a has emerged as a potential therapeutic target for cardiovascular dysfunction and related diseases.
Collapse
Affiliation(s)
- Cun-Cun Hua
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xin-Ming Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Li-Rong Liang
- Department of Clinical Epidemiology and Tobacco Dependence Treatment Research, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Le-Feng Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- *Correspondence: Jiu-Chang Zhong
| | - Jiu-Chang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Tobacco Dependence Treatment Research, Beijing Institute of Respiratory Medicine, Beijing, China
- Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, China
- Le-Feng Wang
| |
Collapse
|
7
|
Yildiz MT, Tutar L, Giritlioğlu NI, Bayram B, Tutar Y. MicroRNAs and Heat Shock Proteins in Breast Cancer Biology. Methods Mol Biol 2022; 2257:293-310. [PMID: 34432285 DOI: 10.1007/978-1-0716-1170-8_15] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Breast cancer has five major immune types; luminal A, luminal B, HER2, Basal-like, and normal-like. Cells produce a family of protein called heat shock proteins (Hsps) in response to exposure to thermal and other proteotoxic stresses play essential roles in cancer metabolism and this large family shows a diverse set of Hsp involvement in different breast cancer immune types. Recently, Hsp members categorized according to their immune type roles. Hsp family consists of several subtypes formed by molecular weight; Hsp70, Hsp90, Hsp100, Hsp40, Hsp60, and small molecule Hsps. Cancer cells employ Hsps as survival factors since most of these proteins prevent apoptosis. Several studies monitored Hsp roles in breast cancer cells and reported Hsp27 involvement in drug resistance, Hsp70 in tumor cell transformation-progression, and interaction with p53. Furthermore, the association of Hsp90 with steroid receptors and signaling proteins in patients with breast cancer directed research to focus on Hsp-based treatments. miRNAs are known to play key roles in all types of cancer that are upregulated or downregulated in cancer which respectively referred to as oncogenes (oncomirs) or tumor suppressors. Expression profiles of miRNAs may be used to classify, diagnose, and predict different cancer types. It is clear that miRNAs play regulatory roles in gene expression and this work reveals miRNA correlation to Hsp depending on specific breast cancer immune types. Deregulation of specific Hsp genes in breast cancer subtypes allows for identification of new targets for drug design and cancer treatment. Here, we performed miRNA network analysis by recruiting Hsp genes detected in breast cancer subtypes and reviewed some of the miRNAs related to aforementioned Hsp genes.
Collapse
Affiliation(s)
- Mehmet Taha Yildiz
- Division of Molecular Medicine, Hamidiye Institute of Health Sciences, University of Health Sciences, Istanbul, Turkey
| | - Lütfi Tutar
- Department of Molecular Biology and Genetics, Faculty of Art and Sciences, Kırşehir Ahi Evran University, Kırşehir, Turkey
| | - Nazlı Irmak Giritlioğlu
- Department of Molecular Medicine, Hamidiye Institute of Health Sciences, University of Health Sciences, Istanbul, Turkey
| | - Banu Bayram
- Department of Nutrition and Dietetics, Hamidiye Faculty of Health Sciences, University of Health Sciences, Istanbul, Turkey
| | - Yusuf Tutar
- Division of Molecular Medicine, Hamidiye Institute of Health Sciences, University of Health Sciences, Istanbul, Turkey.
- Division of Biochemistry, Department of Basic Pharmaceutical Sciences, Hamidiye Faculty of Pharmacy, University of Health Sciences, Istanbul, Turkey.
| |
Collapse
|
8
|
Zang H, Zhang Q, Li X. Non-Coding RNA Networks in Pulmonary Hypertension. Front Genet 2021; 12:703860. [PMID: 34917122 PMCID: PMC8669616 DOI: 10.3389/fgene.2021.703860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 11/08/2021] [Indexed: 01/12/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are involved in various cellular processes. There are several ncRNA classes, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). The detailed roles of these molecules in pulmonary hypertension (PH) remain unclear. We systematically collected and reviewed reports describing the functions of ncRNAs (miRNAs, lncRNAs, and circRNAs) in PH through database retrieval and manual literature reading. The characteristics of identified articles, especially the experimental methods, were carefully reviewed. Furthermore, regulatory networks were constructed using ncRNAs and their interacting RNAs or genes. These data were extracted from studies on pulmonary arterial smooth muscle cells, pulmonary artery endothelial cells, and pulmonary artery fibroblasts. We included 14 lncRNAs, 1 circRNA, 74 miRNAs, and 110 mRNAs in the constructed networks. Using these networks, herein, we describe the current knowledge on the role of ncRNAs in PH. Moreover, these networks actively provide an improved understanding of the roles of ncRNAs in PH. The results of this study are crucial for the clinical application of ncRNAs.
Collapse
Affiliation(s)
- Hongbin Zang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiongyu Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaodong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Salminen A, Kaarniranta K, Kauppinen A. Hypoxia/ischemia impairs CD33 (Siglec-3)/TREM2 signaling: Potential role in Alzheimer's pathogenesis. Neurochem Int 2021; 150:105186. [PMID: 34530055 DOI: 10.1016/j.neuint.2021.105186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/22/2022]
Abstract
Recent genetic and molecular studies have indicated that the innate immune system, especially microglia, have a crucial role in the accumulation of β-amyloid plaques in Alzheimer's disease (AD). In particular, the CD33 receptor, also called Siglec-3, inhibits the TREM2 receptor-induced phagocytic activity of microglia. CD33 receptors recognize the α2,3 and α2,6-linked sialic groups in tissue glycocalyx, especially sialylated gangliosides in human brain. The CD33 receptor triggers cell-type specific responses, e.g., in microglia, CD33 inhibits phagocytosis, whereas in natural killer cells, it inhibits the cytotoxic activity of the NKG2D receptor. Nonetheless, the regulation of the activity of CD33 receptor needs to be clarified. For example, it seems that hypoxia/ischemia, a potential cause of AD pathology, increases the expression of CD33 and its downstream target SHP-1, a tyrosine phosphatase which suppresses the phagocytosis driven by TREM2. Moreover, hypoxia/ischemia increases the deposition of sialylated gangliosides, e.g., GM1, GM2, GM3, and GD1, which are ligands for inhibitory CD33/Siglec-3 receptors. In addition, β-amyloid peptides bind to the sialylated gangliosides in raft-like clusters and subsequently these gangliosides act as seeds for the formation of β-amyloid plaques in AD pathology. It is known that senile plaques contain sialylated GM1, GM2, and GM3 gangliosides, i.e., the same species induced by hypoxia/ischemia treatment. Sialylated gangliosides in plaques might stimulate the CD33/Siglec-3 receptors of microglia and thus impede TREM2-driven phagocytosis. We propose that hypoxia/ischemia, e.g., via the accumulation of sialylated gangliosides, prevents the phagocytosis of β-amyloid deposits by inhibiting CD33/TREM2 signaling.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, FI-70029, KYS, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| |
Collapse
|
10
|
Dou X, Ma Y, Qin Y, Dong Q, Zhang S, Tian R, Pan M. NEAT1 silencing alleviates pulmonary arterial smooth muscle cell migration and proliferation under hypoxia through regulation of miR‑34a‑5p/KLF4 in vitro. Mol Med Rep 2021; 24:749. [PMID: 34468014 DOI: 10.3892/mmr.2021.12389] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/29/2021] [Indexed: 11/06/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe vascular disease that adversely affects patient health and can be life threatening. The present study aimed to investigate the detailed role of nuclear paraspeckle assembly transcript 1 (NEAT1) in PAH. Using RT‑qPCR, the expression levels of NEAT1, microRNA (miR)‑34a‑5p, and Krüppel‑like factor 4 (KLF4) were detected in both hypoxia‑treated pulmonary arterial smooth muscle cells (PASMCs) and serum from PAH patients. Then, the interactions among miR‑34a‑5p, NEAT1, and KLF4 were evaluated by dual‑luciferase reporter assay. The detailed role of the NEAT1/miR‑34a‑5p/KLF4 axis in PAH pathogenesis was further explored using MTT, Transwell, and western blot assays. The results revealed that NEAT1 targeted miR‑34a‑5p and miR‑34a‑5p targeted KLF4. In hypoxia‑treated PASMCs and serum from PAH patients, high NEAT1 and KLF4 expression levels and low miR‑34a‑5p expression were observed. The proliferation and migration of hypoxia‑treated PASMCs were reduced by transfection with sh‑NEAT1 or miR‑34a‑5p mimics. The suppressive effects of NEAT1 knockdown on the proliferation and migration of hypoxia‑treated PASMCs were reversed by knock down of miR‑34a‑5p expression and increased KLF4 expression. NEAT1 was not only highly expressed in the serum of PAH patients but its silencing also alleviated PAH by regulating miR‑34a‑5p/KLF4 in vitro. The present study highlighted a potential new therapeutic target and diagnostic biomarker for PAH.
Collapse
Affiliation(s)
- Xiuli Dou
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Yuxiao Ma
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Yijie Qin
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Qinglin Dong
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Shouwei Zhang
- Department of Medical, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Rui Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Mingyu Pan
- Department of Cardiology, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| |
Collapse
|
11
|
Inhibition of HDAC1 alleviates monocrotaline-induced pulmonary arterial remodeling through up-regulation of miR-34a. Respir Res 2021; 22:239. [PMID: 34465322 PMCID: PMC8408973 DOI: 10.1186/s12931-021-01832-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 08/24/2021] [Indexed: 11/20/2022] Open
Abstract
Background It has been found that up-regulation of histone deacetylases 1 (HDAC1) is involved in the development of pulmonary arterial hypertension (PAH). However, it is still unclear whether inhibition of HDAC1 suppresses the development of PAH via restoring miR-34a level in monocrotaline (MCT)-induced PAH rats. Methods PAH rat models were induced by intraperitoneal injection of MCT. HDAC1 was suppressed by intraperitoneal injection of the class I HDAC inhibitor MS-275, and miR-34a was over-expressed via tail vein injection of miR-34a agomiR. Results HDAC1 protein was significantly increased in MCT-induced PAH rats; this was accompanied with down-regulation of miR-34a and subsequent up-regulation of matrix metalloproteinase 9 (MMP-9)/tissue inhibitor of metalloproteinase 1 (TIMP-1) and MMP-2/TIMP-2. Administration of PAH rats with MS-275 or miR-34a agomiR dramatically abolished MCT-induced reduction of miR-34a and subsequent up-regulation of MMP-9/TIMP-1 and MMP-2/TIMP-2, finally reduced extracellular matrix (ECM) accumulation, pulmonary arterial remodeling, right ventricular systolic pressure (RVSP) and right ventricle hypertrophy index (RVHI) in PAH rats. Conclusions HDAC1 contributes to the development of MCT-induced rat PAH by suppressing miR-34a level and subsequently up-regulating the ratio of MMP-9/TIMP-1 and MMP-2/TIMP-2. Inhibition of HDAC1 alleviates pulmonary arterial remodeling and PAH through up-regulation of miR-34a level and subsequent reduction of MMP-9/TIMP-1 and MMP-2/TIMP-2, suggesting that inhibition of HDAC1 might have potential value in the management of PAH.
Collapse
|
12
|
Zhao M, Wang W, Lu Y, Wang N, Kong D, Shan L. MicroRNA‑153 attenuates hypoxia‑induced excessive proliferation and migration of pulmonary arterial smooth muscle cells by targeting ROCK1 and NFATc3. Mol Med Rep 2021; 23:194. [PMID: 33495839 PMCID: PMC7809904 DOI: 10.3892/mmr.2021.11833] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022] Open
Abstract
The aim of the present study was to explore the effect of microRNA (miR)‑153 on the proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) in a hypoxic condition by targeting ρ‑associated, coiled‑coil‑containing protein kinase 1 (ROCK1) and nuclear factor of activated T cells cytoplasmic 3 (NFATc3). The right ventricular systolic pressure, right ventricular hypertrophy index, medial wall thickness and medial wall area were studied at different time‑points after rats were exposed to hypoxia. Western blot analysis was used to detect ROCK1 and NFATc3 protein levels. In addition, reverse transcription‑quantitative (RT‑q) PCR was performed to confirm the mRNA levels of miR‑153, ROCK1 and NFATc3 in human (H)PASMCs under hypoxic conditions. Transfected cells were then used to evaluate the effect of miR‑153 on cell proliferation and migration abilities. The association between miR‑153 and ROCK1 or NFATc3 was identified through double luciferase assays. Hypoxia induced pulmonary vascular remodeling and pulmonary arterial hypertension, which resulted from the abnormal proliferation of HPASMCs. ROCK1 and NFATc3 were the target genes of miR‑153 and miR‑153 mimic inhibited the protein expressions of ROCK1 and NFATc3 in HPASMCs and further inhibited cell proliferation and migration under hypoxic conditions. By contrast, the miR‑153 inhibitor promoted the proliferation and migration of HPASMCs. miR‑153 regulated the proliferation and migration of HPASMCs under hypoxia by targeting ROCK1 and NFATc3.
Collapse
Affiliation(s)
- Minjie Zhao
- Department of Respiratory Disease, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Wei Wang
- Department of Respiratory Disease, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Ya Lu
- Department of Respiratory Disease, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Nan Wang
- Department of Respiratory Disease, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Delei Kong
- Department of Respiratory Disease, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Lina Shan
- Department of Respiratory Disease, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| |
Collapse
|
13
|
Zhang S, Liu J, Zheng K, Chen L, Sun Y, Yao Z, Sun Y, Lin Y, Lin K, Yuan L. Exosomal miR-211 contributes to pulmonary hypertension via attenuating CaMK1/PPAR-γaxis. Vascul Pharmacol 2021; 136:106820. [PMID: 33238205 DOI: 10.1016/j.vph.2020.106820] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 09/30/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022]
Abstract
AIM Exsomes play a significant role in increasing pathophysiological processes by delivering their content. Recently, a variety of studies have showed exosomal microRNAs (miRNAs) are involved in pulmonary hypertension (PH) notably. In this study, we found that exosomal miR-211 was overexpressed in hypoxia-induced PH rats but its intrinsic regulation was unclear. Therefore, our aim was to reveal the underlying mechanism which overexpressed exosomal miR-211 targeted in the development of PH. METHODS 18 male SD rats were randomly divided into normoxia and hypoxia group, housed in normal or hypoxic chamber for 3 weeks respectively. Then, mean pulmonary arterial pressure (mPAP), pulmonary vascular resistance(PVR), right ventricular hypertrophy index(RV/(LV + S)), the percentage of medial wall area (WA%) and the percentage of medial wall thickness (WT%) were measured. Expression of miR-211 in exosomes was detected by qRT-PCR. Expression of Ca2+/calmodulin-dependent kinase1(CaMK1)and peroxisome proliferator-activated receptors-γ(PPAR-γ)in lung tissue were detected by Western blot(WB); After miR-211 overexpressed exosomes were injected to rats through caudal vein, mPAP, PVR, RV/(LV + S), WA% and WT% were also measured. Sequentially, hypoxia rats were injected with lentivirus riched in miR-211 inhibitor via tail vein, and PH-related indicators were measured. In vitro, after miR-211 was positively or negatively regulated in pulmonary arterial smooth muscle cell (PASMC) by plasmid transfection, proliferation of PASMC was detected by CCK8, as well as the expression of CaMK1 and PPAR- γ. Further, the relationship between CaMK1 and miR-211 was verified by Dual-Luciferase assay. And the regulatory relationship of CaMK1/PPAR- γ aixs was demonstrated in PASMC. RESULTS Evident increases of mPAP, PVR, RVHI, WT% and WA% were observed with hypoxia administration. And the concentration of plasma exosomes in hypoxia rats was increased and positively correlated with the above indexes. miR-211 in exosomes of PH was upregulated while the expression of CaMK1 and PPAR-γ decreased in lung tissues. Further, injection of exosomes overexpressed with miR-211 demonstrated that exosomal miR-211 aggravated PH while inhibition of miR-211 attenuated PH in rats. In vitro, overexpression of miR-211 promoted the proliferation of PASMC and inhibited expression of CaMK1 and PPAR-γ in PASMC. And Dual-luciferase assay demonstrated that CaMK1 was a downstream gene of miR-211. Plasmid transfection experiments indicated that CaMK1 can promote PPAR-γ expression. CONCLUSION Exosomal miR-211 promoted PH via inhibiting CaMK1/PPAR-γ axis, promoting PASMC proliferation in rats.
Collapse
Affiliation(s)
- Shuhao Zhang
- School of First Clinical Medicine, Wenzhou Medical University, Wenzhou, PR China
| | - Jiantao Liu
- School of Second Clinical Medicine, Wenzhou Medical University, Wenzhou, PR China
| | - Kaidi Zheng
- Department of Biochemistry, Basic Medical Science School, Wenzhou Medical University, Wenzhou, PR China
| | - Luowei Chen
- School of First Clinical Medicine, Wenzhou Medical University, Wenzhou, PR China
| | - Yupeng Sun
- School of First Clinical Medicine, Wenzhou Medical University, Wenzhou, PR China
| | - Zhengze Yao
- School of First Clinical Medicine, Wenzhou Medical University, Wenzhou, PR China
| | - Yiruo Sun
- School of Second Clinical Medicine, Wenzhou Medical University, Wenzhou, PR China
| | - Yufan Lin
- School of First Clinical Medicine, Wenzhou Medical University, Wenzhou, PR China
| | - Kexin Lin
- School of Second Clinical Medicine, Wenzhou Medical University, Wenzhou, PR China
| | - Linbo Yuan
- Department of Physiology, Basic Medical Science School, Wenzhou Medical University, Wenzhou, PR China.
| |
Collapse
|
14
|
MicroRNA-34a: the bad guy in age-related vascular diseases. Cell Mol Life Sci 2021; 78:7355-7378. [PMID: 34698884 PMCID: PMC8629897 DOI: 10.1007/s00018-021-03979-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
The age-related vasculature alteration is the prominent risk factor for vascular diseases (VD), namely, atherosclerosis, abdominal aortic aneurysm, vascular calcification (VC) and pulmonary arterial hypertension (PAH). The chronic sterile low-grade inflammation state, alias inflammaging, characterizes elderly people and participates in VD development. MicroRNA34-a (miR-34a) is emerging as an important mediator of inflammaging and VD. miR-34a increases with aging in vessels and induces senescence and the acquisition of the senescence-associated secretory phenotype (SASP) in vascular smooth muscle (VSMCs) and endothelial (ECs) cells. Similarly, other VD risk factors, including dyslipidemia, hyperglycemia and hypertension, modify miR-34a expression to promote vascular senescence and inflammation. miR-34a upregulation causes endothelial dysfunction by affecting ECs nitric oxide bioavailability, adhesion molecules expression and inflammatory cells recruitment. miR-34a-induced senescence facilitates VSMCs osteoblastic switch and VC development in hyperphosphatemia conditions. Conversely, atherogenic and hypoxic stimuli downregulate miR-34a levels and promote VSMCs proliferation and migration during atherosclerosis and PAH. MiR34a genetic ablation or miR-34a inhibition by anti-miR-34a molecules in different experimental models of VD reduce vascular inflammation, senescence and apoptosis through sirtuin 1 Notch1, and B-cell lymphoma 2 modulation. Notably, pleiotropic drugs, like statins, liraglutide and metformin, affect miR-34a expression. Finally, human studies report that miR-34a levels associate to atherosclerosis and diabetes and correlate with inflammatory factors during aging. Herein, we comprehensively review the current knowledge about miR-34a-dependent molecular and cellular mechanisms activated by VD risk factors and highlight the diagnostic and therapeutic potential of modulating its expression in order to reduce inflammaging and VD burn and extend healthy lifespan.
Collapse
|
15
|
Wu Z, Geng J, Qi Y, Li J, Bai Y, Guo Z. MiR-193-3p attenuates the vascular remodeling in pulmonary arterial hypertension by targeting PAK4. Pulm Circ 2020; 10:2045894020974919. [PMID: 33354317 PMCID: PMC7734527 DOI: 10.1177/2045894020974919] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/29/2020] [Indexed: 12/18/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive pulmonary vascular disease associated with dysfunction of pulmonary artery endothelial cells and pulmonary artery smooth muscle cells (PASMCs). To explore the potential mechanism of miR-193-3p in pulmonary arterial hypertension, human PASMCs and rats were respectively stimulated by hypoxia and monocrotaline to establish PAH model in vivo and in vitro. The expressions of miR-193-3p and p21-activated protein kinase 4 (PAK4) in the lung samples of PAH patients and paired healthy samples from the healthy subjects in PHA cells and rats were detected by quantitative reverse transcriptase-PCR. Morphological changes in lung tissues were determined using hematoxylin and eosin staining. Right ventricular systolic pressure (RVSP) and ratio of right ventricle to left ventricle plus septum (RV/LV p S) were measured. The binding relationship between miR-193-3p and PAK4 was analyzed by TargetScan and verified by luciferase reporter assay. Cell viability, apoptosis, and migration were detected by 3-(4, 5-Dimethylthiazol-2- yl)-2,5-diphenyltetrazolium bromide (MTT) flow cytometry, and wound-healing assays, respectively. The protein expressions of PAK4, proliferating cell nuclear antigen (PCNA), P21, p-AKT, and AKT in vivo or in vitro were determined by Western blot. In this study, we found that in pulmonary arterial hypertension, miR-193-3p expression was downregulated and PAK4 expression was up-regulated. MiR-193-3p directly targeted PAK4 and negatively regulated its expression. Hypoxia condition promoted cell proliferation, migration, and inhibited apoptosis accompanied with increased expressions of PCNA and p-AKT/AKT and decreased expression of P21 in PASMCs. MiR-193-3p overexpression attenuated the effects of hypoxia on PASMCs via downregulating PAK4. Monocrotaline treatment increased p-AKT/AKT and decreased P21 expression and caused pulmonary vascular remodeling in the model rats. MiR-193-3p overexpression attenuated pulmonary vascular remodeling, decreased p-AKT/AKT, and increased P21 levels via downregulating PAK4 in monocrotaline-induced rats. The results in this study demonstrated that upregulation of miR-193-3p reduced cell proliferation, migration, and apoptosis of PAH in vitro and pulmonary vascular remodeling in PAH in vivo through downregulating PAK4.
Collapse
Affiliation(s)
- Zhenhua Wu
- Department of Cardiac Surgery, ICU, Tianjin Chest Hospital, Tianjin, China
| | - Jie Geng
- Department of Cardiac Surgery, CICU, Tianjin Chest Hospital, Tianjin, China
| | - Yujuan Qi
- Department of Cardiac Surgery, ICU, Tianjin Chest Hospital, Tianjin, China
| | - Jian Li
- Department of Cardiac Surgery, ICU, Tianjin Chest Hospital, Tianjin, China
| | - Yaobang Bai
- Department of Cardiac Surgery, ICU, Tianjin Chest Hospital, Tianjin, China
| | - Zhigang Guo
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
16
|
Down-regulation of lncRNA Gas5 promotes hypoxia-induced pulmonary arterial smooth muscle cell proliferation by regulating KCNK3 expression. Eur J Pharmacol 2020; 889:173618. [DOI: 10.1016/j.ejphar.2020.173618] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 09/20/2020] [Accepted: 09/29/2020] [Indexed: 01/28/2023]
|
17
|
Wang J, Hu L, Huang H, Yu Y, Wang J, Yu Y, Li K, Li Y, Tian T, Chen F. CAR (CARSKNKDC) Peptide Modified ReNcell-Derived Extracellular Vesicles as a Novel Therapeutic Agent for Targeted Pulmonary Hypertension Therapy. Hypertension 2020; 76:1147-1160. [DOI: 10.1161/hypertensionaha.120.15554] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent years, mesenchymal stem cells (MSCs)–derived extracellular vesicles (EVs) are emerging as a potential therapeutic agent for pulmonary hypertension (PH). However, the full realization of MSCs-derived EVs therapy has been hampered by the absence of standardization in MSCs culture and the challenges of industrial scale-up. The study was to exploit an alternative replacement for MSCs using currently commercialized stem cell lines for effective targeted PH therapy. ReNcell VM—a human neural stem cell line—has been utilized here as a reliable and easily adoptable source of EVs. We first demonstrated that ReNcell-derived EVs (ReNcell-EVs) pretreatment effectively prevented Su/Hx (SU5416/hypoxia)-induced PH in mice. Then for targeted therapy, we conjugated ReNcell-EVs with CAR (CARSKNKDC) peptide (CAR-EVs)—a peptide identified to specifically target hypertensive pulmonary arteries, by bio-orthogonal chemistry. Intravenous administration of CAR-EVs selectively targeted hypertensive pulmonary artery lesions especially pulmonary artery smooth muscle cells. Moreover, compared with unmodified ReNcell-EVs, CAR-EVs treatment significantly improved therapeutic effect in reversing Su/Hx-induced PH in mice. Mechanistically, ReNcell-EVs inhibited hypoxia-induced proliferation, migration, and phenotype switch of pulmonary artery smooth muscle cells, at least in part, via the delivery of its endogenous highly expressed miRNAs, let-7b-5p, miR-92b-3p, and miR-100-5p. In addition, we also found that ReNcell-EVs inhibited hypoxia-induced cell apoptosis and endothelial-mesenchymal transition in human microvascular endothelial cells. Taken together, our results provide an alternative to MSCs-derived EVs–based PH therapy via using ReNcell as a reliable source of EVs. Particularly, our CAR-conjugated EVs may serve as a novel drug carrier that enhances the specificity and efficiency of drug delivery for effective PH-targeted therapy.
Collapse
Affiliation(s)
- Jie Wang
- From the Department of Forensic Medicine (Jie Wang, L.H., H.H., Yanfang Yu, Youjia Yu, K.L., Y.L., F.C.), Nanjing Medical University, Jiangsu, China
| | - Li Hu
- From the Department of Forensic Medicine (Jie Wang, L.H., H.H., Yanfang Yu, Youjia Yu, K.L., Y.L., F.C.), Nanjing Medical University, Jiangsu, China
| | - Huijie Huang
- From the Department of Forensic Medicine (Jie Wang, L.H., H.H., Yanfang Yu, Youjia Yu, K.L., Y.L., F.C.), Nanjing Medical University, Jiangsu, China
| | - Yanfang Yu
- From the Department of Forensic Medicine (Jie Wang, L.H., H.H., Yanfang Yu, Youjia Yu, K.L., Y.L., F.C.), Nanjing Medical University, Jiangsu, China
| | - Jingshen Wang
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu (Jingshen Wang, T.T.), Nanjing Medical University, Jiangsu, China
| | - Youjia Yu
- From the Department of Forensic Medicine (Jie Wang, L.H., H.H., Yanfang Yu, Youjia Yu, K.L., Y.L., F.C.), Nanjing Medical University, Jiangsu, China
| | - Kai Li
- From the Department of Forensic Medicine (Jie Wang, L.H., H.H., Yanfang Yu, Youjia Yu, K.L., Y.L., F.C.), Nanjing Medical University, Jiangsu, China
| | - Yan Li
- From the Department of Forensic Medicine (Jie Wang, L.H., H.H., Yanfang Yu, Youjia Yu, K.L., Y.L., F.C.), Nanjing Medical University, Jiangsu, China
| | - Tian Tian
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu (Jingshen Wang, T.T.), Nanjing Medical University, Jiangsu, China
| | - Feng Chen
- From the Department of Forensic Medicine (Jie Wang, L.H., H.H., Yanfang Yu, Youjia Yu, K.L., Y.L., F.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine (F.C.), Nanjing Medical University, Jiangsu, China
| |
Collapse
|
18
|
Bogner EM, Daly AF, Gulde S, Karhu A, Irmler M, Beckers J, Mohr H, Beckers A, Pellegata NS. miR-34a is upregulated in AIP-mutated somatotropinomas and promotes octreotide resistance. Int J Cancer 2020; 147:3523-3538. [PMID: 32856736 DOI: 10.1002/ijc.33268] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 07/15/2020] [Accepted: 08/12/2020] [Indexed: 12/17/2022]
Abstract
Pituitary adenomas (PAs) are intracranial tumors associated with significant morbidity due to hormonal dysregulation, mass effects and have a heavy treatment burden. Growth hormone (GH)-secreting PAs (somatotropinomas) cause acromegaly-gigantism. Genetic forms of somatotropinomas due to germline AIP mutations (AIPmut+) have an early onset and are aggressive and resistant to treatment with somatostatin analogs (SSAs), including octreotide. The molecular underpinnings of these clinical features remain unclear. We investigated the role of miRNA dysregulation in AIPmut+ vs AIPmut- PA samples by array analysis. miR-34a and miR-145 were highly expressed in AIPmut+ vs AIPmut- somatotropinomas. Ectopic expression of AIPmut (p.R271W) in Aip-/- mouse embryonic fibroblasts (MEFs) upregulated miR-34a and miR-145, establishing a causal link between AIPmut and miRNA expression. In PA cells (GH3), miR-34a overexpression promoted proliferation, clonogenicity, migration and suppressed apoptosis, whereas miR-145 moderately affected proliferation and apoptosis. Moreover, high miR-34a expression increased intracellular cAMP, a critical mitogenic factor in PAs. Crucially, high miR-34a expression significantly blunted octreotide-mediated GH inhibition and antiproliferative effects. miR-34a directly targets Gnai2 encoding Gαi2, a G protein subunit inhibiting cAMP production. Accordingly, Gαi2 levels were significantly lower in AIPmut+ vs AIPmut- PA. Taken together, somatotropinomas with AIP mutations overexpress miR-34a, which in turn downregulates Gαi2 expression, increases cAMP concentration and ultimately promotes cell growth. Upregulation of miR-34a also impairs the hormonal and antiproliferative response of PA cells to octreotide. Thus, miR-34a is a novel downstream target of mutant AIP that promotes a cellular phenotype mirroring the aggressive clinical features of AIPmut+ acromegaly.
Collapse
Affiliation(s)
- Eva-Maria Bogner
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| | - Adrian F Daly
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, Liège Université, Liège, Belgium
| | - Sebastian Gulde
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| | - Auli Karhu
- Department of Medical and Clinical Genetics & Genome-Scale Biology Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Technische Universität München, Chair of Experimental Genetics, Freising, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Hermine Mohr
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| | - Albert Beckers
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, Liège Université, Liège, Belgium
| | - Natalia S Pellegata
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
19
|
Tang HX, Qin XP, Li J. Role of the signal transducer and activator of transcription 3 protein in the proliferation of vascular smooth muscle cells. Vascular 2020; 28:821-828. [PMID: 32486969 DOI: 10.1177/1708538120929504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Cardiovascular disease (CVD) remains the primary cause of morbidity and mortality worldwide. The abnormal proliferation of vascular smooth muscle cells (VSMCs) is a key event in the pathogenesis of CVD. The functional and phenotypic changes in vascular cells are mediated by complex signaling cascades that initiate and control genetic reprogramming. Many studies have demonstrated that signal transducer and activator of transcription 3 (STAT3) regulates a diverse array of functions relevant to atherosclerosis. METHODS In this review, we summarize the studies on the STAT3-mediated proliferation of VSMCs and subsequent CVDs such as hypertension, atherosclerosis, stroke, coronary artery disease, and myocardial infarction. Furthermore, we describe the general background of STAT3, its structure, function and regulation as well as the STAT3 signaling pathway. Finally, we highlight some potential issues and propose some solutions to these issues.Results and conclusions: STAT3 activation promotes the proliferation of VSMCs by regulating the transcription of genes. Studying the mechanism of VSMC proliferation induced by the STAT3 pathway is valuable for finding therapeutic targets for CVD.
Collapse
Affiliation(s)
- Hong-Xia Tang
- The First People's Hospital of Chenzhou, Institute of Pharmacy and Pharmacology, University of South China, Hunan, China
| | - Xu-Ping Qin
- The First People's Hospital of Chenzhou, Institute of Pharmacy and Pharmacology, University of South China, Hunan, China
| | - Jie Li
- The First People's Hospital of Chenzhou, Institute of Pharmacy and Pharmacology, University of South China, Hunan, China
- School of Pharmacy, Southern Medical University, Guangdong, China
| |
Collapse
|
20
|
Lai YT, Chao HW, Lai ACY, Lin SH, Chang YJ, Huang YS. CPEB2-activated PDGFRα mRNA translation contributes to myofibroblast proliferation and pulmonary alveologenesis. J Biomed Sci 2020; 27:52. [PMID: 32295602 PMCID: PMC7160907 DOI: 10.1186/s12929-020-00643-0] [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: 11/24/2019] [Accepted: 03/26/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alveologenesis is the final stage of lung development to form air-exchanging units between alveoli and blood vessels. Genetic susceptibility or hyperoxic stress to perturb this complicated process can cause abnormal enlargement of alveoli and lead to bronchopulmonary dysplasia (BPD)-associated emphysema. Platelet-derived growth factor receptor α (PDGFRα) signaling is crucial for alveolar myofibroblast (MYF) proliferation and its deficiency is associated with risk of BPD, but posttranscriptional mechanisms regulating PDGFRα synthesis during lung development remain largely unexplored. Cytoplasmic polyadenylation element-binding protein 2 (CPEB2) is a sequence-specific RNA-binding protein and translational regulator. Because CPEB2-knockout (KO) mice showed emphysematous phenotypes, we investigated how CPEB2-controlled translation affects pulmonary development and function. METHODS Respiratory and pulmonary functions were measured by whole-body and invasive plethysmography. Histological staining and immunohistochemistry were used to analyze morphology, proliferation, apoptosis and cell densities from postnatal to adult lungs. Western blotting, RNA-immunoprecipitation, reporter assay, primary MYF culture and ectopic expression rescue were performed to demonstrate the role of CPEB2 in PDGFRα mRNA translation and MYF proliferation. RESULTS Adult CPEB2-KO mice showed emphysema-like dysfunction. The alveolar structure in CPEB2-deficient lungs appeared normal at birth but became simplified through the alveolar stage of lung development. In CPEB2-null mice, we found reduced proliferation of MYF progenitors during alveolarization, abnormal deposition of elastin and failure of alveolar septum formation, thereby leading to enlarged pulmonary alveoli. We identified that CPEB2 promoted PDGFRα mRNA translation in MYF progenitors and this positive regulation could be disrupted by H2O2, a hyperoxia-mimetic treatment. Moreover, decreased proliferating ability in KO MYFs due to insufficient PDGFRα expression was rescued by ectopic expression of CPEB2, suggesting an important role of CPEB2 in upregulating PDGFRα signaling for pulmonary alveologenesis. CONCLUSIONS CPEB2-controlled translation, in part through promoting PDGFRα expression, is indispensable for lung development and function. Since defective pulmonary PDGFR signaling is a key feature of human BPD, CPEB2 may be a risk factor for BPD.
Collapse
Affiliation(s)
- Yen-Ting Lai
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd, Taipei, 11529, Taiwan
| | - Hsu-Wen Chao
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Alan Chuan-Ying Lai
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd, Taipei, 11529, Taiwan
| | - Shu-Hui Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ya-Jen Chang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd, Taipei, 11529, Taiwan.
| | - Yi-Shuian Huang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd, Taipei, 11529, Taiwan.
| |
Collapse
|
21
|
Zhang J, Li Y, Qi J, Yu X, Ren H, Zhao X, Xin W, He S, Zheng X, Ma C, Zhang L, Wu B, Zhu D. Circ- calm4 Serves as an miR-337-3p Sponge to Regulate Myo10 (Myosin 10) and Promote Pulmonary Artery Smooth Muscle Proliferation. Hypertension 2020; 75:668-679. [PMID: 32008463 DOI: 10.1161/hypertensionaha.119.13715] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Pulmonary artery smooth muscle cell proliferation is the pathological basis of pulmonary vascular remodeling in hypoxic pulmonary hypertension. Recent studies suggest that circular RNA (circRNA) can regulate various biological processes, including cell proliferation. Therefore, it is possible that circRNA may have important roles in pulmonary artery smooth muscle cell proliferation in hypoxic pulmonary hypertension. In the present study, we aimed to identify functional circRNAs and clarify their roles and mechanisms in pulmonary artery smooth muscle cell proliferation in pulmonary hypertension. RNA sequencing identified 67 circRNAs that were differentially expressed in hypoxic lung tissues of mice. Screening by bioinformatics and quantitative polymerase chain reaction revealed significant elevation of a circRNA derived from alternative splicing of the calmodulin 4 gene (designated circ-calm4). Notably, this circRNA absorbed miR-337-3p. We further identified Myo10 (myosin 10) as a target protein of miR-337-3p. miR-337-3p bound to the 3'-untranslated region of Myo10 mRNA, thereby attenuating the translation of Myo10. Using loss-of-function and gain-of-function approaches, we found that circ-calm4 regulated cell proliferation by regulating the cell cycle. Additionally, we verified the functions of miR-337-3p and Myo10 in hypoxic pulmonary artery smooth muscle. Our results suggested that the circ-calm4/miR-337-3p/Myo10 signal transduction axis modulated the proliferation of pulmonary artery smooth muscle cells at the molecular level, thus establishing potential targets for the early diagnosis and treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Junting Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X. Zhao, L.Z., C.M., D.Z.), Harbin Medical University (Daqing), China.,Department of Pharmacology, College of Pharmacy (J.Z., Y.L., J.Q., H.R.,W.X., S.H., D.Z.), Harbin Medical University (Daqing), China.,Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China
| | - Yiying Li
- Department of Pharmacology, College of Pharmacy (J.Z., Y.L., J.Q., H.R.,W.X., S.H., D.Z.), Harbin Medical University (Daqing), China.,Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China
| | - Jing Qi
- Department of Pharmacology, College of Pharmacy (J.Z., Y.L., J.Q., H.R.,W.X., S.H., D.Z.), Harbin Medical University (Daqing), China.,Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China
| | - Xiufeng Yu
- Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China
| | - Huanhuan Ren
- Department of Pharmacology, College of Pharmacy (J.Z., Y.L., J.Q., H.R.,W.X., S.H., D.Z.), Harbin Medical University (Daqing), China.,Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China
| | - Xijuan Zhao
- From the College of Medical Laboratory Science and Technology (X.Y., X. Zhao, L.Z., C.M., D.Z.), Harbin Medical University (Daqing), China.,Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China
| | - Wei Xin
- Department of Pharmacology, College of Pharmacy (J.Z., Y.L., J.Q., H.R.,W.X., S.H., D.Z.), Harbin Medical University (Daqing), China.,Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China
| | - Siyu He
- Department of Pharmacology, College of Pharmacy (J.Z., Y.L., J.Q., H.R.,W.X., S.H., D.Z.), Harbin Medical University (Daqing), China.,Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China
| | - Xiaodong Zheng
- Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China
| | - Cui Ma
- From the College of Medical Laboratory Science and Technology (X.Y., X. Zhao, L.Z., C.M., D.Z.), Harbin Medical University (Daqing), China.,Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China
| | - Lixin Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X. Zhao, L.Z., C.M., D.Z.), Harbin Medical University (Daqing), China.,Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China
| | - Bingxiang Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Harbin Medical University, China (B.W.)
| | - Daling Zhu
- From the College of Medical Laboratory Science and Technology (X.Y., X. Zhao, L.Z., C.M., D.Z.), Harbin Medical University (Daqing), China.,Department of Pharmacology, College of Pharmacy (J.Z., Y.L., J.Q., H.R.,W.X., S.H., D.Z.), Harbin Medical University (Daqing), China.,Central Laboratory of Harbin Medical University (Daqing), China (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., C.M., L.Z., D.Z.).,College of Pharmacy (J.Z., Y.L., J.Q., X.Y., H.R., X. Zhao, W.X., S.H., X. Zheng, C.M., L.Z., D.Z.), Harbin Medical University, China.,Key Laboratory of Cardiovascular Medicine Research, Ministry of Education (D.Z.), Harbin Medical University, China.,State Province Key Laboratories of Biomedicine-Pharmaceutics of China (D.Z.)
| |
Collapse
|
22
|
Bisserier M, Janostiak R, Lezoualc’h F, Hadri L. Targeting epigenetic mechanisms as an emerging therapeutic strategy in pulmonary hypertension disease. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2020; 2:R17-R34. [PMID: 32161845 PMCID: PMC7065685 DOI: 10.1530/vb-19-0030] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial cardiopulmonary disease characterized by an elevation of pulmonary artery pressure (PAP) and pulmonary vascular resistance (PVR), which can lead to right ventricular (RV) failure, multi-organ dysfunction, and ultimately to premature death. Despite the advances in molecular biology, the mechanisms underlying pulmonary hypertension (PH) remain unclear. Nowadays, there is no curative treatment for treating PH. Therefore, it is crucial to identify novel, specific therapeutic targets and to offer more effective treatments against the progression of PH. Increasing amounts of evidence suggest that epigenetic modification may play a critical role in the pathogenesis of PAH. In the presented paper, we provide an overview of the epigenetic mechanisms specifically, DNA methylation, histone acetylation, histone methylation, and ncRNAs. As the recent identification of new pharmacological drugs targeting these epigenetic mechanisms has opened new therapeutic avenues, we also discuss the importance of epigenetic-based therapies in the context of PH.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Radoslav Janostiak
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Frank Lezoualc’h
- Inserm, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, University of Toulouse, Toulouse Cedex 4, France
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
23
|
MicroRNA-802 induces hepatitis B virus replication and replication through regulating SMARCE1 expression in hepatocellular carcinoma. Cell Death Dis 2019; 10:783. [PMID: 31611549 PMCID: PMC6791889 DOI: 10.1038/s41419-019-1999-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 08/22/2019] [Accepted: 08/26/2019] [Indexed: 12/17/2022]
Abstract
Growing evidences have indicated that microRNAs (miRNAs) can regulate hepatitis B virus (HBV) expression and replication, playing crucial roles in the development of HBV infection. Until now, the functional role and mechanism of miR-802 in HBV replication and expression remain unknown. We indicated that miR-802 expression was upregulated in the HBV-associated hepatocellular carcinoma (HCC) tissues compared with the adjacent noncancerous samples. In addition, we showed that the SMARCE1 expression level was downregulated in the HBV-associated HCC tissues compared with the adjacent noncancerous samples. miR-802 expression was negatively related with MARCE1 expression in HBV-associated HCC tissues. Moreover, miR-802 expression was upregulated, and SMARCE1 expression was downregulated in the HBV-infected HepG2.2.15 cells. Ectopic expression of miR-802 significantly enhanced HBV DNA replication, while knockdown of miR-802 significantly decreased HBV DNA replication. We showed that overexpression of miR-802 promoted HbsAg and HbeAg expression, while inhibition of miR-802 decreased HbsAg and HbeAg expression. Furthermore, we indicated that ectopic expression of SMARCE1 suppressed HBV DNA replication and decreased the expression level of HbsAg and HbeAg. Finally, we showed that overexpression of miR-802 promoted HBV DNA replication through regulating SMARCE1 expression. These results suggested the important roles of miR-802 on HBV expression and replication, which may shed new light on the development of treatment for HBV.
Collapse
|
24
|
The neuroprotective effects of carvacrol on ischemia/reperfusion-induced hippocampal neuronal impairment by ferroptosis mitigation. Life Sci 2019; 235:116795. [PMID: 31470002 DOI: 10.1016/j.lfs.2019.116795] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/26/2019] [Accepted: 08/26/2019] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Cerebral ischemia is the most common type of neuronal injury and is characterized by a reduction in the function and number of hippocampal neurons. Carvacrol has a significant neuroprotective effect in cerebral ischemia. However, the mechanisms by which carvacrol affects cerebral ischemia, especially with respect to the regulation of neuronal damage by iron levels, have never been systematically studied. This study aimed to reveal the mechanisms by which carvacrol protects against hippocampal neuron impairment after ischemic stroke in gerbils. MATERIALS AND METHODS The Morris water maze test was performed to evaluate learning and memory impairments. Iron ion content and oxidative stress index were detected by the kit. MTT assay was performed to assess the cell viability. The morphology and molecular characteristics were detected by electron micrographs and western blot. RESULTS In the present study, we demonstrated the neuroprotective effects of carvacrol in vivo and in vitro. The Morris water maze test showed that the learning and memory abilities of the gerbils treated with carvacrol were significantly improved. Lipid peroxide injury was evaluated by measuring the levels of lipid peroxide biomarkers; the results indicated that carvacrol decreased the level of lipid peroxide in ischemic gerbil brain tissue. Histopathological examinations and western blotting were performed to evaluate injury in neurons, and carvacrol reduced cell death. Moreover, ferroptosis in the hippocampus was evaluated by measuring the levels of proteins involved in this iron-dependent form of regulated cell death. These results indicated that carvacrol reduced cell death and that carvacrol inhibited ferroptosis by increasing the expression of glutathione peroxidase 4(GPx4). This study showed that carvacrol may be a valuable drug for treating cerebral ischemia. CONCLUSION Carvacrol provides protection for hippocampal neurons against I/R in gerbils by inhibiting ferroptosis through increasing the expression of GPx4.
Collapse
|
25
|
Chen S, Yu C, Lu R, Song T, Wang X, Tang W, Gao Y. miR-107 inhibits PDGF-BB-induced proliferation of human pulmonary arterial smooth muscle cells and migration through targeting NOR1. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1599-1608. [PMID: 31933977 PMCID: PMC6947117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/25/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Abnormal proliferation of PASMCs is the main phenotype of pulmonary arterial hypertension (PAH). MicroRNAs (miRNAs) were reported to participate in regulating the progression of PAH. Here, we aimed to investigate the impact of miR-107 on proliferation and migration of PASMCs and potential mechanism. METHODS MTT assay was carried out to examine the cell viability of PASMCs. PASMC migration ability was verified through Transwell assay. RT-qPCR was performed to detect the expression of miR-107 and NOR1. Western blot was conducted to detect the expression of cell proliferation markers Ki-67, p27 and Cyclin D1, as well as NOR1. Bioinformatics analysis was conducted to verify whether the 3'-untranslated region (3'-UTR) of NOR1 contains a binding site for miR-107, and luciferase reporter assay and RNA immunoprecipitation (RIP) were employed to confirm the relationship between miR-107 and NOR1. RESULTS Platelet-derived growth factor (PDGF)-BB promoted the cell viability and migration of PASMCs, and suppressed miR-107 expression in a time-dependent and concentration-dependent manner. Introduction of miR-107 inhibited the promotion of proliferation and migration of PASMCs stimulated by PDGF-BB, while loss of miR-107 facilitated PDGF-BB-induced promoted effects. NOR1 was identified as a downstream gene of miR-107 and down-regulated by miR-107. Knockout of NOR1 also repressed the promotion of proliferation and migration of PASMCs stimulated by PDGF-BB. Additionally, restoration of NOR1 attenuated the inhibition of miR-107 on the cell viability and migration ability of PASMCs. CONCLUSION miR-107 inhibits PDGF-BB-induced PASMCs proliferation and migration through targeting NOR1.
Collapse
Affiliation(s)
- Shiyuan Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College Changhuai Road 287, Bengbu 233003, China
| | - Chaowen Yu
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College Changhuai Road 287, Bengbu 233003, China
| | - Ran Lu
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College Changhuai Road 287, Bengbu 233003, China
| | - Tao Song
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College Changhuai Road 287, Bengbu 233003, China
| | - Xiaogao Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College Changhuai Road 287, Bengbu 233003, China
| | - Wenbo Tang
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College Changhuai Road 287, Bengbu 233003, China
| | - Yong Gao
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College Changhuai Road 287, Bengbu 233003, China
| |
Collapse
|
26
|
MiR-34b-5p Mediates the Proliferation and Differentiation of Myoblasts by Targeting IGFBP2. Cells 2019; 8:cells8040360. [PMID: 30999686 PMCID: PMC6523632 DOI: 10.3390/cells8040360] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/11/2019] [Accepted: 04/14/2019] [Indexed: 12/12/2022] Open
Abstract
As key post-transcriptional regulators, microRNAs (miRNAs) play an indispensable role in skeletal muscle development. Our previous study suggested that miR-34b-5p and IGFBP2 could have a potential role in skeletal muscle growth. Our goal in this study is to explore the function and regulatory mechanism of miR-34b-5p and IGFBP2 in myogenesis. In this study, the dual-luciferase reporter assay and Western blot analysis showed that IGFBP2 is a direct target of miR-34b-5p. Flow cytometric analysis and EdU assay showed that miR-34b-5p could repress the cell cycle progression of myoblasts, and miR-34b-5p could promote the formation of myotubes by promoting the expression of MyHC. On the contrary, the overexpression of IGFBP2 significantly facilitated the proliferation of myoblasts and hampered the formation of myotubes. Together, our results indicate that miR-34b-5p could mediate the proliferation and differentiation of myoblasts by targeting IGFBP2.
Collapse
|
27
|
Li Z, Ma J, Bi J, Guo H, Chan MTV, Wu WKK, Wu Z, Shen J. MicroRNA signature of air pollution exposure‐induced congenital defects. J Cell Physiol 2019; 234:17896-17904. [PMID: 30883755 DOI: 10.1002/jcp.28422] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/10/2019] [Accepted: 02/14/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Zheng Li
- Department of Orthopaedic Surgery Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jianqing Ma
- Department of Orthopedic Surgery The General Hospital of Xingtai Mining Industry Bloc., Orthopaedic Hospital of Xingtai, Xingtai Hebei China
| | - Jiaqi Bi
- Department of Orthopaedic Surgery Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Haiwei Guo
- Department of Orthopaedic Surgery Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Matthew T. V. Chan
- Department of Anaesthesia and Intensive Care The Chinese University of Hong Kong Hong Kong China
| | - William K. K. Wu
- Department of Anaesthesia and Intensive Care The Chinese University of Hong Kong Hong Kong China
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong Hong Kong China
| | - Zhanyong Wu
- Department of Orthopedic Surgery The General Hospital of Xingtai Mining Industry Bloc., Orthopaedic Hospital of Xingtai, Xingtai Hebei China
| | - Jianxiong Shen
- Department of Orthopaedic Surgery Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| |
Collapse
|
28
|
Rippe C, Albinsson S, Guron G, Nilsson H, Swärd K. Targeting transcriptional control of soluble guanylyl cyclase via NOTCH for prevention of cardiovascular disease. Acta Physiol (Oxf) 2019; 225:e13094. [PMID: 29754438 DOI: 10.1111/apha.13094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/04/2018] [Accepted: 05/04/2018] [Indexed: 12/18/2022]
Abstract
Soluble guanylyl cyclase (sGC) is an effector enzyme of nitric oxide (NO). Recent work has unravelled how levels of this enzyme are controlled, and highlighted a role in vascular disease. We provide a timely summary of available knowledge on transcriptional regulation of sGC, including influences from the NOTCH signalling pathway and genetic variants. It is speculated that hypertension-induced repression of sGC starts a vicious circle that can be initiated by periods of stress, diet or genetic factors, and a key tenet is that reduction in sGC further raises blood pressure. The idea that dysregulation of sGC contributes to syndromes caused by defective NOTCH signalling is advanced, and we discuss drug repositioning for vascular disease prevention. The advantage of targeting sGC expression rather than activity is also considered. It is argued that transcriptional inputs on sGC arise from interactions with other cells, the extracellular matrix and microRNAs (miRNAs), and concluded that the promise of sGC as a target for prevention of cardiovascular disease has increased in recent time.
Collapse
Affiliation(s)
- C. Rippe
- Department of Experimental Medical Science; Lund University; Lund Sweden
| | - S. Albinsson
- Department of Experimental Medical Science; Lund University; Lund Sweden
| | - G. Guron
- Department of Physiology; University of Gothenburg; Gothenburg Sweden
| | - H. Nilsson
- Department of Physiology; University of Gothenburg; Gothenburg Sweden
| | - K. Swärd
- Department of Experimental Medical Science; Lund University; Lund Sweden
| |
Collapse
|
29
|
Fang B, Wei L, Dong K, Niu X, Sui X, Zhang H. miR-202 modulates the progression of neuropathic pain through targeting RAP1A. J Cell Biochem 2018; 120:2973-2982. [PMID: 30520098 DOI: 10.1002/jcb.27025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/05/2018] [Indexed: 12/16/2022]
Abstract
Neuropathic pain is a somatosensory disorder which is caused by disease or nerve injury that affects the nervous system. microRNAs (miRNAs) are proved to play crucial roles in the development of neuropathic pain. However, the role of miR-202 in neuropathic pain is still unknown. Sprague-Dawley rats were used for constructing the neuropathic pain model. The expression of miR-202 was determined by quantitative real-time polymerase chain reaction. Potential target gene for miR-202 was measured using bioinformatics methods and Western blot analysis. In this study, we used rats to establish a neuropathic pain model and measured the effect of miR-202 in neuropathic pain. We demonstrated that miR-202 expression was downregulated in the spinal dorsal horn of bilateral sciatic nerve chronic constriction injury (bCCI) rat. However, miR-202 expression was not changed in the dorsal root ganglion, hippocampus, and anterior cingulated cortex of bCCI rat. We identified that RAP1A was a direct target gene of miR-202 in the PC12 cell. RAP1A expression was upregulated in the spinal dorsal horn of bCCI rat. Overexpression of miR-202 could improve the pain threshold for bCCI rats in both hindpaws, indicating that miR-202 overexpression could lighten the pain threshold for model rats. Moreover, RAP1A overexpression increased the pain threshold effect of miR-202 overexpression treated bCCI rats, indicating that miR-202 could lighten the pain threshold through inhibiting RAP1A expression. These data suggested that miR-202 acted pivotal roles in the development of neuropathic pain partly through targeting RAP1A gene.
Collapse
Affiliation(s)
- Baojun Fang
- Department of Anestheiology, Dongchangfu District Maternal and Child Health Hospital, Liaocheng, Shandong, China
| | - Limin Wei
- Department of Anestheiology, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Kejun Dong
- Department of Anestheiology, Rizhao People's Hospital, Rizhao, Shandong, China
| | - Xiaohui Niu
- Department of Anestheiology, Lingcheng People's Hospital, Dezhou, Shandong, China
| | - Xiuhui Sui
- Tranditional Chinese Medicine Hospital of Gaotang, Liaocheng, Shandong, China
| | - Hongquan Zhang
- Department of Anestheiology, Liaocheng People's Hospital, Liaocheng, Shandong, China
| |
Collapse
|
30
|
Efremov YR, Proskurina AS, Potter EA, Dolgova EV, Efremova OV, Taranov OS, Ostanin AA, Chernykh ER, Kolchanov NA, Bogachev SS. Cancer Stem Cells: Emergent Nature of Tumor Emergency. Front Genet 2018; 9:544. [PMID: 30505319 PMCID: PMC6250818 DOI: 10.3389/fgene.2018.00544] [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: 07/09/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
A functional analysis of 167 genes overexpressed in Krebs-2 tumor initiating cells was performed. In the first part of the study, the genes were analyzed for their belonging to one or more of the three groups, which represent the three major phenotypic manifestation of malignancy of cancer cells, namely (1) proliferative self-sufficiency, (2) invasive growth and metastasis, and (3) multiple drug resistance. 96 genes out of 167 were identified as possible contributors to at least one of these fundamental properties. It was also found that substantial part of these genes are also known as genes responsible for formation and/or maintenance of the stemness of normal pluri-/multipotent stem cells. These results suggest that the malignancy is simply the ability to maintain the stem cell specific genes expression profile, and, as a consequence, the stemness itself regardless of the controlling effect of stem niches. In the second part of the study, three stress factors combined into the single concept of "generalized cellular stress," which are assumed to activate the expression of these genes, were defined. In addition, possible mechanisms for such activation were identified. The data obtained suggest the existence of a mechanism for the de novo formation of a pluripotent/stem phenotype in the subpopulation of "committed" tumor cells.
Collapse
Affiliation(s)
- Yaroslav R Efremov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Anastasia S Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Ekaterina A Potter
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgenia V Dolgova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Oksana V Efremova
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Oleg S Taranov
- The State Research Center of Virology and Biotechnology Vector, Koltsovo, Russia
| | - Aleksandr A Ostanin
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Elena R Chernykh
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Nikolay A Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Sergey S Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
31
|
Bourgeois A, Omura J, Habbout K, Bonnet S, Boucherat O. Pulmonary arterial hypertension: New pathophysiological insights and emerging therapeutic targets. Int J Biochem Cell Biol 2018; 104:9-13. [DOI: 10.1016/j.biocel.2018.08.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/24/2018] [Accepted: 08/29/2018] [Indexed: 12/17/2022]
|
32
|
Johnson JL. Elucidating the contributory role of microRNA to cardiovascular diseases (a review). Vascul Pharmacol 2018; 114:31-48. [PMID: 30389614 PMCID: PMC6445803 DOI: 10.1016/j.vph.2018.10.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/13/2018] [Accepted: 10/28/2018] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases encompassing atherosclerosis, aortic aneurysms, restenosis, and pulmonary arterial hypertension, remain the leading cause of morbidity and mortality worldwide. In response to a range of stimuli, the dynamic interplay between biochemical and biomechanical mechanisms affect the behaviour and function of multiple cell types, driving the development and progression of cardiovascular diseases. Accumulating evidence has highlighted microRNAs (miRs) as significant regulators and micro-managers of key cellular and molecular pathophysiological processes involved in predominant cardiovascular diseases, including cell mitosis, motility and viability, lipid metabolism, generation of inflammatory mediators, and dysregulated proteolysis. Human pathological and clinical studies have aimed to identify select microRNA which may serve as biomarkers of disease and their progression, which are discussed within this review. In addition, I provide comprehensive coverage of in vivo investigations elucidating the modulation of distinct microRNA on the pathophysiology of atherosclerosis, abdominal aortic aneurysms, restenosis, and pulmonary arterial hypertension. Collectively, clinical and animal studies have begun to unravel the complex and often diverse effects microRNAs and their targets impart during the development of cardiovascular diseases and revealed promising therapeutic strategies through which modulation of microRNA function may be applied clinically.
Collapse
Affiliation(s)
- Jason L Johnson
- Laboratory of Cardiovascular Pathology, Bristol Medical School, University of Bristol, UK.
| |
Collapse
|
33
|
Miao C, Chang J, Zhang G. Recent research progress of microRNAs in hypertension pathogenesis, with a focus on the roles of miRNAs in pulmonary arterial hypertension. Mol Biol Rep 2018; 45:2883-2896. [DOI: 10.1007/s11033-018-4335-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/28/2018] [Indexed: 12/22/2022]
|
34
|
Luo X, Li J, Yin L, Pan J, Zhang Y, Jiang Z. Role of microRNA 146a on the healing of cornea alkali burn treated with mesenchymal stem cells. Mol Med Rep 2018; 18:3203-3210. [PMID: 30066863 PMCID: PMC6102633 DOI: 10.3892/mmr.2018.9328] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 06/09/2017] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to investigate the effect of microRNA 146a (miR146a) on promoting the repair of corneal alkali burn with bone marrow mesenchymal stem cells (MSCs). A total of 24 Sprague‑Dawley female rats were divided into a normal group (Control), a normal MSC treatment group (Normal MSCs), an miR146a knockout MSC treatment group (miR146a‑low MSCs) and an miR146a high‑expression MSC treatment group (miR146a‑high MSCs) according to the random number table. Quantitative polymerase chain reaction was used to evaluate the expression levels of miR146a. MTT assay was performed to measure the cell viability of mesenchymal stem cells (MSCs) and apoptosis was measured by flow cytometry. The expression levels of p65 nuclear factor (NF)‑κB, proliferating cell nuclear antigen (PCNA) and Fas proteins were analyzed by western blotting. MSCs were tested for the secretion levels of vascular endothelial growth factor (VEGF), CD45, interferon (IFN)‑γ and interleukin (IL)‑10 by ELISA. The miR146a‑high MSCs improved cell viability of MSCs and inhibited apoptosis of MSCs following alkali burn. miR146a‑high MSCs decreased the expression levels of p65NF‑κB and PCNA, and enhanced the expression level of Fas. Furthermore, miR146a‑high MSCs improved the cornea opacity and enhanced the inhibition of neovascularization in the rats following alkali burn. miR146a‑high MSCs inhibit the expression of VEGF, CD45, IFN‑γ, while enhanced the expression of IL‑10. Therefore, miR146a promotes the repair of corneal alkali burn in rats treated with MSCs.
Collapse
Affiliation(s)
- Xu Luo
- Department of Wounds and Burns, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jianmin Li
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Lihui Yin
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jian Pan
- Department of Ophthalmology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yang Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zipei Jiang
- Department of Ophthalmology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
35
|
Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu XQ, Zhang L. Gestational Hypoxia and Developmental Plasticity. Physiol Rev 2018; 98:1241-1334. [PMID: 29717932 PMCID: PMC6088145 DOI: 10.1152/physrev.00043.2017] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hypoxia is one of the most common and severe challenges to the maintenance of homeostasis. Oxygen sensing is a property of all tissues, and the response to hypoxia is multidimensional involving complicated intracellular networks concerned with the transduction of hypoxia-induced responses. Of all the stresses to which the fetus and newborn infant are subjected, perhaps the most important and clinically relevant is that of hypoxia. Hypoxia during gestation impacts both the mother and fetal development through interactions with an individual's genetic traits acquired over multiple generations by natural selection and changes in gene expression patterns by altering the epigenetic code. Changes in the epigenome determine "genomic plasticity," i.e., the ability of genes to be differentially expressed according to environmental cues. The genomic plasticity defined by epigenomic mechanisms including DNA methylation, histone modifications, and noncoding RNAs during development is the mechanistic substrate for phenotypic programming that determines physiological response and risk for healthy or deleterious outcomes. This review explores the impact of gestational hypoxia on maternal health and fetal development, and epigenetic mechanisms of developmental plasticity with emphasis on the uteroplacental circulation, heart development, cerebral circulation, pulmonary development, and the hypothalamic-pituitary-adrenal axis and adipose tissue. The complex molecular and epigenetic interactions that may impact an individual's physiology and developmental programming of health and disease later in life are discussed.
Collapse
Affiliation(s)
- Charles A. Ducsay
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Ravi Goyal
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - William J. Pearce
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Xiang-Qun Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
36
|
Tian X, Yu C, Shi L, Li D, Chen X, Xia D, Zhou J, Xu W, Ma C, Gu L, An Y. MicroRNA-199a-5p aggravates primary hypertension by damaging vascular endothelial cells through inhibition of autophagy and promotion of apoptosis. Exp Ther Med 2018; 16:595-602. [PMID: 30116316 PMCID: PMC6090226 DOI: 10.3892/etm.2018.6252] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 04/13/2018] [Indexed: 12/19/2022] Open
Abstract
The present study investigated the expression of microRNA (miRNA or miR)-199a-5p in the peripheral blood of patients with primary hypertension, and examined its mechanism of action in vascular endothelial cell injury induced by hypertension. A total of 57 patients with primary hypertension, who were treated at the Affiliated Hospital of Qingdao University (Qingdao, China) between December 2014 and November 2015 were included in the present study. Peripheral blood was collected from all patients. The expression of miR-199a-5p was measured using reverse-transcription quantitative polymerase chain reaction analysis. Human umbilical vein endothelial cells (HUVECs) were divided into negative control, miR-199a-5p mimics and rescue (co-transfected with miR-199a-5p mimics and inhibitor) groups. After transfection, the proliferation and apoptosis of HUVECs were evaluated by a Cell Counting Kit-8 assay, a bromodeoxyuridine incorporation assay and flow cytometry. Western blot analysis was used to determine the expression of proteins involved in autophagy-associated and adenosine monophosphate kinase (AMPK)/unc-51 like autophagy activating kinase 1 (ULK1) signaling pathways. Laser scanning confocal microscopy and electron microscopy were used to observe the autophagy of HUVECs. The expression of miR-199a-5p was elevated in peripheral blood of patients with hypertension, and was correlated with the progression of hypertension. Overexpression of miR-199a-5p inhibited the proliferation and promoted the apoptosis of HUVECs. Upon expression of miR-199a-5p, the transition between microtubule-associated proteins 1A/1B light chain 3B (LC3B)I and LC3BII proteins was inhibited, the expression of p62 protein was upregulated. In addition, miR-199a-5p decreased the numbers of autophagosomes and autolysosomes in HUVECs. The present study demonstrated that expression of miR-199a-5p is positively correlated with the severity of hypertension. Expression of miR-199a-5p aggravated vascular endothelial injury by inhibiting autophagy and promoting the apoptosis of HUVECs via downregulation of the AMPK/ULK1 signaling pathway.
Collapse
Affiliation(s)
- Xintao Tian
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Chunpeng Yu
- Department of Intervention, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Lei Shi
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Dan Li
- Department of Cardiovascular Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Xiaoxue Chen
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Di Xia
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Jingwei Zhou
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Wanqun Xu
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Chengtai Ma
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Lihua Gu
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Yi An
- Department of Cardiovascular Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| |
Collapse
|
37
|
Xu N, Yang W, Liu Y, Yan F, Yu Z. MicroRNA-411 promoted the osteosarcoma progression by suppressing MTSS1 expression. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:12064-12071. [PMID: 29453719 DOI: 10.1007/s11356-018-1331-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 01/18/2018] [Indexed: 06/08/2023]
Abstract
MicroRNAs (miRNAs) play crucial roles in the progression of different tumors. In our study, we investigated the expression and roles of miR-411 in human osteosarcoma. In this study, we first confirmed that the miR-411 expression was higher in the serum of patients with osteosarcoma than in the serum of healthy volunteers. In addition, we found that the miR-411 expression was upregulated in the osteosarcoma tissues compared to that in the matched normal bone tissues. We also demonstrated that the miR-411 expression was upregulated in the four osteosarcoma cell lines. Elevated expression of miR-411 promoted osteosarcoma cell proliferation and migration. Moreover, we identified that metastasis suppressor protein 1 (MTSS1) was a direct target gene of miR-411 in the osteosarcoma cell. We also demonstrated that the MTSS1 expression was downregulated in the osteosarcoma tissues compared to that in the matched normal bone tissues. In addition, MTSS1 expression level was inversely correlated with miR-411 expression in the osteosarcoma tissues. Furthermore, elevated expression of miR-411 enhanced the osteosarcoma cell proliferation and migration through inhibiting the MTSS1 expression. These data suggested that miR-411 played as oncogene in the osteosarcoma partly by inhibiting the MTSS1 expression.
Collapse
Affiliation(s)
- Ning Xu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng St, Nangang, Harbin, Heilongjiang, China
| | - Weiliang Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng St, Nangang, Harbin, Heilongjiang, China
| | - Yang Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng St, Nangang, Harbin, Heilongjiang, China
| | - Feng Yan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng St, Nangang, Harbin, Heilongjiang, China
| | - Zhange Yu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng St, Nangang, Harbin, Heilongjiang, China.
| |
Collapse
|
38
|
Chen KH, Dasgupta A, Lin J, Potus F, Bonnet S, Iremonger J, Fu J, Mewburn J, Wu D, Dunham-Snary K, Theilmann AL, Jing ZC, Hindmarch C, Ormiston ML, Lawrie A, Archer SL. Epigenetic Dysregulation of the Dynamin-Related Protein 1 Binding Partners MiD49 and MiD51 Increases Mitotic Mitochondrial Fission and Promotes Pulmonary Arterial Hypertension: Mechanistic and Therapeutic Implications. Circulation 2018; 138:287-304. [PMID: 29431643 DOI: 10.1161/circulationaha.117.031258] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/22/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Mitotic fission is increased in pulmonary arterial hypertension (PAH), a hyperproliferative, apoptosis-resistant disease. The fission mediator dynamin-related protein 1 (Drp1) must complex with adaptor proteins to cause fission. Drp1-induced fission has been therapeutically targeted in experimental PAH. Here, we examine the role of 2 recently discovered, poorly understood Drp1 adapter proteins, mitochondrial dynamics protein of 49 and 51 kDa (MiD49 and MiD51), in normal vascular cells and explore their dysregulation in PAH. METHODS Immunoblots of pulmonary artery smooth muscle cells (control, n=6; PAH, n=8) and immunohistochemistry of lung sections (control, n=6; PAH, n=6) were used to assess the expression of MiD49 and MiD51. The effects of manipulating MiDs on cell proliferation, cell cycle, and apoptosis were assessed in human and rodent PAH pulmonary artery smooth muscle cells with flow cytometry. Mitochondrial fission was studied by confocal imaging. A microRNA (miR) involved in the regulation of MiD expression was identified using microarray techniques and in silico analyses. The expression of circulatory miR was assessed with quantitative reverse transcription-polymerase chain reaction in healthy volunteers (HVs) versus patients with PAH from Sheffield, UK (plasma: HV, n=29, PAH, n=27; whole blood: HV, n=11, PAH, n=14) and then confirmed in a cohort from Beijing, China (plasma: HV, n=19, PAH, n=36; whole blood: HV, n=20, PAH, n=39). This work was replicated in monocrotaline and Sugen 5416-hypoxia, preclinical PAH models. Small interfering RNAs targeting MiDs or an miR mimic were nebulized to rats with monocrotaline-induced PAH (n=4-10). RESULTS MiD expression is increased in PAH pulmonary artery smooth muscle cells, which accelerates Drp1-mediated mitotic fission, increases cell proliferation, and decreases apoptosis. Silencing MiDs (but not other Drp1 binding partners, fission 1 or mitochondrial fission factor) promotes mitochondrial fusion and causes G1-phase cell cycle arrest through extracellular signal-regulated kinases 1/2- and cyclin-dependent kinase 4-dependent mechanisms. Augmenting MiDs in normal cells causes fission and recapitulates the PAH phenotype. MiD upregulation results from decreased miR-34a-3p expression. Circulatory miR-34a-3p expression is decreased in both patients with PAH and preclinical models of PAH. Silencing MiDs or augmenting miR-34a-3p regresses experimental PAH. CONCLUSIONS In health, MiDs regulate Drp1-mediated fission, whereas in disease, epigenetic upregulation of MiDs increases mitotic fission, which drives pathological proliferation and apoptosis resistance. The miR-34a-3p-MiD pathway offers new therapeutic targets for PAH.
Collapse
Affiliation(s)
- Kuang-Hueih Chen
- Department of Medicine (K.-H.C., A.D., F.P., J.F., J.M., D.W., K.D.-S., A.L.T., M.L.O., S.L.A.)
| | - Asish Dasgupta
- Department of Medicine (K.-H.C., A.D., F.P., J.F., J.M., D.W., K.D.-S., A.L.T., M.L.O., S.L.A.)
| | - Jianhui Lin
- Queen's University, Kingston, Canada. Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK (J.L., J.I, A.L.)
| | - François Potus
- Department of Medicine (K.-H.C., A.D., F.P., J.F., J.M., D.W., K.D.-S., A.L.T., M.L.O., S.L.A.)
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group of the University Cardiology and Pulmonary Institute of the Quebec Research Centre, Laval University, Quebec City, Canada (S.B.)
| | - James Iremonger
- Queen's University, Kingston, Canada. Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK (J.L., J.I, A.L.)
| | - Jennifer Fu
- Department of Medicine (K.-H.C., A.D., F.P., J.F., J.M., D.W., K.D.-S., A.L.T., M.L.O., S.L.A.)
| | - Jeffrey Mewburn
- Department of Medicine (K.-H.C., A.D., F.P., J.F., J.M., D.W., K.D.-S., A.L.T., M.L.O., S.L.A.).,Queen's Cardiopulmonary Unit, Translational Institute of Medicine, Department of Medicine (J.M., C.H., M.L.O., S.L.A.)
| | - Danchen Wu
- Department of Medicine (K.-H.C., A.D., F.P., J.F., J.M., D.W., K.D.-S., A.L.T., M.L.O., S.L.A.)
| | - Kimberly Dunham-Snary
- Department of Medicine (K.-H.C., A.D., F.P., J.F., J.M., D.W., K.D.-S., A.L.T., M.L.O., S.L.A.)
| | - Anne L Theilmann
- Department of Medicine (K.-H.C., A.D., F.P., J.F., J.M., D.W., K.D.-S., A.L.T., M.L.O., S.L.A.)
| | - Zhi-Cheng Jing
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Z.-C.J.)
| | - Charles Hindmarch
- Queen's Cardiopulmonary Unit, Translational Institute of Medicine, Department of Medicine (J.M., C.H., M.L.O., S.L.A.)
| | - Mark L Ormiston
- Department of Medicine (K.-H.C., A.D., F.P., J.F., J.M., D.W., K.D.-S., A.L.T., M.L.O., S.L.A.).,Queen's Cardiopulmonary Unit, Translational Institute of Medicine, Department of Medicine (J.M., C.H., M.L.O., S.L.A.)
| | - Allan Lawrie
- Queen's University, Kingston, Canada. Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK (J.L., J.I, A.L.)
| | - Stephen L Archer
- Department of Medicine (K.-H.C., A.D., F.P., J.F., J.M., D.W., K.D.-S., A.L.T., M.L.O., S.L.A.) .,Queen's Cardiopulmonary Unit, Translational Institute of Medicine, Department of Medicine (J.M., C.H., M.L.O., S.L.A.)
| |
Collapse
|
39
|
Effect of miR-29b on the Proliferation and Apoptosis of Pulmonary Artery Smooth Muscle Cells by Targeting Mcl-1 and CCND2. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6051407. [PMID: 29662889 PMCID: PMC5831881 DOI: 10.1155/2018/6051407] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 12/11/2017] [Indexed: 12/20/2022]
Abstract
The proliferation and apoptosis of pulmonary artery smooth muscle cells (PASMCs) are considered to be key steps in the progression of pulmonary arterial hypertension (PAH). MicroRNAs (e.g., miR-29b) have been identified in various diseases to be critical modulators of cell growth and apoptosis by targeting Mcl-1 and CCND2. However, the role of miR-29b in PAH remains unknown. So we try to investigate the effect of miR-29b on Mcl-1 and CCND2 protein in PASMCs, analyze the effect of miR-29b on the proliferation of PASMCs, and explore the significance of miR-29b in the proliferation, apoptosis, and gene therapy of PAH. It was observed that gene chip analysis showed miR-29b expression in pulmonary artery tissue. The expression of miR-29b was significantly reduced in PAH model mice. MiR-29b inhibited the proliferation of PASMCs and promoted the apoptosis of PASMCs. Mechanically, miR-29b could inhibit the expression of Mcl-1 and CCND2 protein and silenced Mcl-1 and CCND2 could abolish the change of proliferation and apoptosis of PASMCs. These results demonstrate that miR-29b suppressed cellular proliferation and promoted apoptosis of PASMCs, possibly through the inhibition of Mcl-1 and CCND2. Therefore, miR-29b may serve as a useful therapeutic tool to treat PAH.
Collapse
|
40
|
Gangwar RS, Rajagopalan S, Natarajan R, Deiuliis JA. Noncoding RNAs in Cardiovascular Disease: Pathological Relevance and Emerging Role as Biomarkers and Therapeutics. Am J Hypertens 2018; 31:150-165. [PMID: 29186297 DOI: 10.1093/ajh/hpx197] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022] Open
Abstract
Noncoding RNAs (ncRNA) include a diverse range of functional RNA species-microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) being most studied in pathophysiology. Cardiovascular morbidity is associated with differential expression of myriad miRNAs; miR-21, miR-155, miR-126, miR-146a/b, miR-143/145, miR-223, and miR-221 are the top 9 most reported miRNAs in hypertension and atherosclerotic disease. A single miRNA may have hundreds of messenger RNA targets, which makes a full appreciation of the physiologic ramifications of such broad-ranging effects a challenge. miR-21 is the most prominent ncRNA associated with hypertension and atherosclerotic disease due to its role as a "mechano-miR", responding to arterial shear stresses. "Immuno-miRs", such as miR-155 and miR-223, affect cardiovascular disease (CVD) via regulation of hematopoietic cell differentiation, chemotaxis, and activation in response to many pro-atherogenic stimuli. "Myo-miRs", such as miR-1 and miR-133, affect cardiac muscle plasticity and remodeling in response to mechanical overload. This in-depth review analyzes observational and experimental reports of ncRNAs in CVD, including future applications of ncRNA-based strategies in diagnosis, prediction (e.g., survival and response to small molecule therapy), and biologic therapy.
Collapse
Affiliation(s)
- Roopesh S Gangwar
- Cardiovascular Research Institute (CVRI), Case Western Reserve University, Cleveland, Ohio, USA
| | - Sanjay Rajagopalan
- Cardiovascular Research Institute (CVRI), Case Western Reserve University, Cleveland, Ohio, USA
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Jeffrey A Deiuliis
- Cardiovascular Research Institute (CVRI), Case Western Reserve University, Cleveland, Ohio, USA
- Department of Medicine, Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
41
|
Tang Z, Qiu H, Luo L, Liu N, Zhong J, Kang K, Gou D. miR-34b Modulates Skeletal Muscle Cell Proliferation and Differentiation. J Cell Biochem 2017; 118:4285-4295. [PMID: 28422320 DOI: 10.1002/jcb.26079] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/18/2017] [Indexed: 12/22/2022]
Abstract
Myogenesis involves myoblast proliferation and differentiation to myocytes, followed by fusion and hypertrophy to form myotubes during muscle development. Increasing evidence showed that microRNAs (miRNAs) play important roles in the regulation of myogenesis. We have previously revealed that miR-34b is steadily increased during this process. This miRNA regulates differentiation in various cell types, though its function in myogenesis remains to be elucidated. In this study, we show that miR-34b represses muscle cell proliferation and promotes myotube formation. Our quantitative iTRAQ-based proteomic analysis reveals 97 proteins are regulated by miR-34b in mouse myoblast C2C12. We identified that miR-34b targets 14-3-3 protein gamma, adenosylhomocysteinase and nucleolin by binding to their 3'UTR. Further analysis of these proteins expression patterns show that nucleolin is a cognate target of miR-34b during myogenic differentiation. Here, we proved that a moderate reduction of nucleolin in cells enhanced the myotube formation. However, nucleolin is required for myogenesis, as cells with low levels of nucleolin reduced cell proliferation rate and are unable to differentiate. Our data demonstrated that nucleolin regulates myogenesis in a protein-abundance-dependent manner. J. Cell. Biochem. 118: 4285-4295, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zhixiong Tang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Huiling Qiu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
- Biomedical Engineering, Health and Environmental Engineering, Shenzhen Technology University, Shenzhen, Guangdong, 518000, China
| | - Lan Luo
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Nian Liu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Jiasheng Zhong
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Kang Kang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University Health Sciences Center, Shenzhen, 518000, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| |
Collapse
|
42
|
PDGFRα Regulated by miR-34a and FoxO1 Promotes Adipogenesis in Porcine Intramuscular Preadipocytes through Erk Signaling Pathway. Int J Mol Sci 2017; 18:ijms18112424. [PMID: 29140299 PMCID: PMC5713392 DOI: 10.3390/ijms18112424] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 11/04/2017] [Accepted: 11/11/2017] [Indexed: 12/27/2022] Open
Abstract
Suitable intramuscular fat (IMF) content improves porcine meat quality. The vital genes regulating IMF deposition are necessary for the selection and breeding of an IMF trait. However, the effect and mechanism of PDGFRα on IMF deposition are still unclear. Here, PDGFRα is moderately expressed in porcine longissimus dorsi muscle (LD), whereas it highly expressed in white adipose tissue (WAT). Moreover, PDGFRα-positive cells were located in the gaps of LD fibers which there were IMF adipocytes. Compared with 180-day-old and lean-type pigs, the levels of PDGFRα were much higher in one-day-old and fat-type pigs. Meanwhile the levels of PDGFRα gradually decreased during IMF preadipocyte differentiation. Furthermore, PDGFRα promoted adipogenic differentiation through activating Erk signaling pathway. Based on PDGFRα upstream regulation analysis, we found that the knockdown of FoxO1 repressed lipogenesis by downregulating PDGFRα, and miR-34a inhibited adipogenesis through targeting PDGFRα. Collectively, PDGFRα is a positive regulator of IMF deposition. Therefore, we suggest that PDGFRα is a possible target to improve meat quality.
Collapse
|
43
|
Consales C, Cirotti C, Filomeni G, Panatta M, Butera A, Merla C, Lopresto V, Pinto R, Marino C, Benassi B. Fifty-Hertz Magnetic Field Affects the Epigenetic Modulation of the miR-34b/c in Neuronal Cells. Mol Neurobiol 2017; 55:5698-5714. [PMID: 29039021 DOI: 10.1007/s12035-017-0791-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/26/2017] [Indexed: 12/24/2022]
Abstract
The exposure to extremely low-frequency magnetic fields (ELF-MFs) has been associated to increased risk of neurodegenerative diseases, although the underlying molecular mechanisms are still undefined. Since epigenetic modulation has been recently encountered among the key events leading to neuronal degeneration, we here aimed at assessing if the control of gene expression mediated by miRNAs, namely miRs-34, has any roles in driving neuronal cell response to 50-Hz (1 mT) magnetic field in vitro. We demonstrate that ELF-MFs drive an early reduction of the expression level of miR-34b and miR-34c in SH-SY5Y human neuroblastoma cells, as well as in mouse primary cortical neurons, by affecting the transcription of the common pri-miR-34. This modulation is not p53 dependent, but attributable to the hyper-methylation of the CpG island mapping within the miR-34b/c promoter. Incubation with N-acetyl-l-cysteine or glutathione ethyl-ester fails to restore miR-34b/c expression, suggesting that miRs-34 are not responsive to ELF-MF-induced oxidative stress. By contrast, we show that miRs-34 control reactive oxygen species production and affect mitochondrial oxidative stress triggered by ELF-MFs, likely by modulating mitochondria-related miR-34 targets identified by in silico analysis. We finally demonstrate that ELF-MFs alter the expression of the α-synuclein, which is specifically stimulated upon ELF-MFs exposure via both direct miR-34 targeting and oxidative stress. Altogether, our data highlight the potential of the ELF-MFs to tune redox homeostasis and epigenetic control of gene expression in vitro and shed light on the possible mechanism(s) producing detrimental effects and predisposing neurons to degeneration.
Collapse
Affiliation(s)
- Claudia Consales
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.
| | - Claudia Cirotti
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy.,Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Martina Panatta
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Alessio Butera
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Caterina Merla
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.,Vectorology and Anticancer Therapies, UMR 8203, CNRS, Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Vanni Lopresto
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Rosanna Pinto
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Carmela Marino
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Barbara Benassi
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.
| |
Collapse
|
44
|
Pan J, Li K, Huang W, Zhang X. MiR-137 inhibited cell proliferation and migration of vascular smooth muscle cells via targeting IGFBP-5 and modulating the mTOR/STAT3 signaling. PLoS One 2017; 12:e0186245. [PMID: 29016699 PMCID: PMC5634643 DOI: 10.1371/journal.pone.0186245] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 09/27/2017] [Indexed: 12/28/2022] Open
Abstract
Abnormal proliferation of vascular smooth muscle cells (VSMCs) contributes to the development of cardiovascular diseases. Studies have shown the great impact of microRNAs (miRNAs) on the cell proliferation of VSMCs. This study examined the effects of miR-137 on the cell proliferation and migration of VSMCs and also explored the underlying molecular mechanisms. The mRNA and protein expression levels were determined by qRT-PCR and western blot assays, respectively. The CCK-8 assay, wound healing assay and transwell migration assay were performed to measure cell proliferation and migration of VSMCs. The miR-137-targeted 3’untranslated region of insulin-like growth factor-binding protein-5 (IGFBP-5) was confirmed by luciferase reporter assay. Platelet-derived growth factor-bb (PDGF-bb) treatment enhanced cell proliferation and suppressed the expression of miR-137 in VSMCs. The gain-of-function and loss-of-function assays showed that overexpression of miR-137 suppressed the cell proliferation and migration, and also inhibited the expression of matrix genes of VSMCs; down-regulation of miR-137 had the opposite effects on VSMCs. Bioinformatics analysis and luciferase report assay results showed that IGFBP-5 was a direct target of miR-137, and miR-137 overexpression suppressed the IGFBP-5 expression and down-regulation of miR-137 increased the IGFBP-5 expression in VSMCs. PDGF-bb treatment also increased the IGFBP-5 mRNA expression. In addition, enforced expression of IGFBP-5 reversed the inhibitory effects of miR-137 on cell proliferation and migration of VSMCs. More importantly, overexpression of miR-137 also suppressed the activity of mTOR/STAT3 signaling in VSMCs. Taken together, the results suggest that miR-137 may suppress cell proliferation and migration of VSMCs via targeting IGFBP-5 and modulating mTOR/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Jin Pan
- Clinical Medical College, Xi'an Medical University, Xi'an City, Shaanxi Province, China
- * E-mail:
| | - Kai Li
- Department of Cardiology, the First Affiliated Hospital of Xi'an Medical University, Xi'an City, Shaanxi Province, China
| | - Wei Huang
- Clinical Medical College, Xi'an Medical University, Xi'an City, Shaanxi Province, China
| | - Xiaoqing Zhang
- Clinical Medical College, Xi'an Medical University, Xi'an City, Shaanxi Province, China
| |
Collapse
|
45
|
Chen D, Gao W, Wang S, Ni B, Gao Y. Critical effects of epigenetic regulation in pulmonary arterial hypertension. Cell Mol Life Sci 2017; 74:3789-3808. [PMID: 28573430 PMCID: PMC11107652 DOI: 10.1007/s00018-017-2551-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 05/14/2017] [Accepted: 05/29/2017] [Indexed: 12/11/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by persistent pulmonary vasoconstriction and pulmonary vascular remodeling. The pathogenic mechanisms of PAH remain to be fully clarified and measures of effective prevention are lacking. Recent studies; however, have indicated that epigenetic processes may exert pivotal influences on PAH pathogenesis. In this review, we summarize the latest research findings regarding epigenetic regulation in PAH, focusing on the roles of non-coding RNAs, histone modifications, ATP-dependent chromatin remodeling and DNA methylation, and discuss the potential of epigenetic-based therapies for PAH.
Collapse
Affiliation(s)
- Dewei Chen
- Department of Pathophysiology and High Altitude Pathology, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, People's Republic of China
- Key Laboratory of High Altitude Medicine of PLA, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Wenxiang Gao
- Department of Pathophysiology and High Altitude Pathology, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, People's Republic of China
- Key Laboratory of High Altitude Medicine of PLA, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Shouxian Wang
- Department of Pathophysiology and High Altitude Pathology, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, People's Republic of China
- Key Laboratory of High Altitude Medicine of PLA, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Bing Ni
- Department of Pathophysiology and High Altitude Pathology, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, People's Republic of China.
- Key Laboratory of High Altitude Medicine of PLA, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, 400038, People's Republic of China.
| | - Yuqi Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, People's Republic of China.
- Key Laboratory of High Altitude Medicine of PLA, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
46
|
Jiang R, Shi Y, Zeng C, Yu W, Zhang A, Du Y. Protein kinase Cα stimulates hypoxia‑induced pulmonary artery smooth muscle cell proliferation in rats through activating the extracellular signal‑regulated kinase 1/2 pathway. Mol Med Rep 2017; 16:6814-6820. [PMID: 28901444 PMCID: PMC5865839 DOI: 10.3892/mmr.2017.7478] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 07/18/2017] [Indexed: 01/11/2023] Open
Abstract
Hypoxic pulmonary hypertension (HPH) may contribute to vascular remodeling, and pulmonary artery smooth muscle cell (PASMC) proliferation has an important role in this process. However, no relevant information concerning the role and mechanism of protein kinase C (PKC)α in hypoxia-induced rat PASMC proliferation has been elucidated. The present study aimed to further investigate this by comparison of rat PASMC proliferation among normoxia for 72 h (21% O2), hypoxia for 72 h (3% O2), hypoxia + promoter 12-myristate 13-acetate control, hypoxia + safingol control, hypoxia + PD98059 control and hypoxia + U0126 control groups. The present study demonstrated that protein expression levels of PKCα in rat PASMCs were elevated. In conclusion, through activating the extracellular signal-regulated 1/2 signaling pathway, PKCα is involved in and initiates PASMC proliferation, thus bringing about pulmonary artery hypertension. These results add to the understanding of the mechanism PKCα in PH formation and lays a theoretical basis for prevention as well as treatment of HPH.
Collapse
Affiliation(s)
- Rui Jiang
- Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yiwei Shi
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, Taiyuan, Shanxi 030001, P.R. China
| | - Chao Zeng
- Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Wenyan Yu
- Respiratory Department, Central Hospital of Zibo, Zibo, Shandong 255036, P.R. China
| | - Aizhen Zhang
- Department of Respiratory and Critical Care Medicine, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030012, P.R. China
| | - Yongcheng Du
- Department of Respiratory and Critical Care Medicine, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030012, P.R. China
| |
Collapse
|
47
|
Jin C, Feng Y, Ni Y, Shan Z. MicroRNA-610 suppresses osteosarcoma oncogenicity via targeting TWIST1 expression. Oncotarget 2017; 8:56174-56184. [PMID: 28915582 PMCID: PMC5593553 DOI: 10.18632/oncotarget.17045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/24/2017] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma is the most frequent primary bone tumor affects adolescents and young adults. Recently, microRNAs (miRNAs) are short, non-coding and endogenous RNAs that played as important roles in the initiation and progression of tumors. In this study, we try to explore the biological function and expression of miR-610 in the osteosarcoma. We showed that miR-610 expression was downregulated in the osteosarcoma tissues and cell lines. Elevated expression of miR-610 suppressed the osteosarcoma cell proliferation, cell cycle, invasion and EMT program. Moreover, overexpression of miR-610 increased sensitivity of MG-63 and U2OS cells to cisplatin. Twist1 was identified as a direct target gene of miR-610 in the osteosarcoma cell. Furthermore, we demonstrated that Twist1 was upregulated in the osteosarcoma tissues and cell lines. The expression of Twist1 was negatively associated with the expression of miR-610 expression in the osteosarcoma tissues. Ectopic expression of Twist1 inhibited the sensitivity of miR-610-overexpressing MG-63 cells to cisplatin. We also showed that overexpression of Twist1 increased the proliferation and invasion of miR-610-overexpressing MG-63 cells. These data indicated that ectopic expression of miR-610 suppressed the osteosarcoma cell proliferation, cell cylce, invasion and increased the sensitivity of osteosarcoma cells to cisplatin through targeting the Twist1 expression.
Collapse
Affiliation(s)
- Chi Jin
- The Third Department of Orthopaedics, Central Hospital of Cangzhou City, Cangzhou, Hebei, China
| | - Yongjian Feng
- The Fourth Department of Orthopaedics, Central Hospital of Cangzhou City, Cangzhou, Hebei, China
| | - Yongjian Ni
- The Third Department of Orthopaedics, Central Hospital of Cangzhou City, Cangzhou, Hebei, China
| | - Zhonglin Shan
- The Third Department of Orthopaedics, Central Hospital of Cangzhou City, Cangzhou, Hebei, China
| |
Collapse
|
48
|
Abstract
Pulmonary hypertension (PH) is a multifaceted vascular disease where development and severity are determined by both genetic and environmental factors. Over the past decade, there has been an acceleration of the discovery of molecular effectors that mediate PH pathogenesis, including large numbers of microRNA molecules that are expressed in pulmonary vascular cell types and exert system-wide regulatory functions in all aspects of vascular health and disease. Due to the inherent pleiotropy, overlap, and redundancy of these molecules, it has been challenging to define their integrated effects on overall disease manifestation. In this review, we summarize our current understanding of the roles of microRNAs in PH with an emphasis on potential methods to discern the hierarchical motifs governing their multifunctional and interconnected activities. Deciphering this higher order of regulatory structure will be crucial for overcoming the challenges of developing these molecules as biomarkers or therapeutic targets, in isolation or combination.
Collapse
|
49
|
MicroRNA-34 directly targets pair-rule genes and cytoskeleton component in the honey bee. Sci Rep 2017; 7:40884. [PMID: 28098233 PMCID: PMC5241629 DOI: 10.1038/srep40884] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 12/12/2016] [Indexed: 01/06/2023] Open
Abstract
MicroRNAs (miRNAs) are key regulators of developmental processes, such as cell fate determination and differentiation. Previous studies showed Dicer knockdown in honeybee embryos disrupt the processing of functional mature miRNAs and impairs embryo patterning. Here we investigated the expression profiles of miRNAs in honeybee embryogenesis and the role of the highly conserved miR-34-5p in the regulation of genes involved in insect segmentation. A total of 221 miRNAs were expressed in honey bee embryogenesis among which 97 mature miRNA sequences have not been observed before. Interestingly, we observed a switch in dominance between the 5-prime and 3-prime arm of some miRNAs in different embryonic stages; however, most miRNAs present one dominant arm across all stages of embryogenesis. Our genome-wide analysis of putative miRNA-target networks and functional pathways indicates miR-34-5p is one of the most conserved and connected miRNAs associated with the regulation of genes involved in embryonic patterning and development. In addition, we experimentally validated that miR-34-5p directly interacts to regulatory elements in the 3'-untranslated regions of pair-rule (even-skipped, hairy, fushi-tarazu transcription factor 1) and cytoskeleton (actin5C) genes. Our study suggests that miR-34-5p may regulate the expression of pair-rule and cytoskeleton genes during early development and control insect segmentation.
Collapse
|
50
|
Martinez-Sanchez A, Rutter GA, Latreille M. MiRNAs in β-Cell Development, Identity, and Disease. Front Genet 2017; 7:226. [PMID: 28123396 PMCID: PMC5225124 DOI: 10.3389/fgene.2016.00226] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/21/2016] [Indexed: 12/22/2022] Open
Abstract
Pancreatic β-cells regulate glucose metabolism by secreting insulin, which in turn stimulates the utilization or storage of the sugar by peripheral tissues. Insulin insufficiency and a prolonged period of insulin resistance are usually the core components of type 2 diabetes (T2D). Although, decreased insulin levels in T2D have long been attributed to a decrease in β-cell function and/or mass, this model has recently been refined with the recognition that a loss of β-cell “identity” and dedifferentiation also contribute to the decline in insulin production. MicroRNAs (miRNAs) are key regulatory molecules that display tissue-specific expression patterns and maintain the differentiated state of somatic cells. During the past few years, great strides have been made in understanding how miRNA circuits impact β-cell identity. Here, we review current knowledge on the role of miRNAs in regulating the acquisition of the β-cell fate during development and in maintaining mature β-cell identity and function during stress situations such as obesity, pregnancy, aging, or diabetes. We also discuss how miRNA function could be harnessed to improve our ability to generate β-cells for replacement therapy for T2D.
Collapse
Affiliation(s)
- Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London London, UK
| | - Mathieu Latreille
- Cellular Identity and Metabolism Group, MRC London Institute of Medical SciencesLondon, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College LondonLondon, UK
| |
Collapse
|