1
|
Bardill JR, Karimpour-Fard A, Breckenfelder CC, Sucharov CC, Eason CR, Gallagher LT, Khailova L, Wright CJ, Gien J, Galan HL, Derderian SC. microRNAs in congenital diaphragmatic hernia: insights into prenatal and perinatal biomarkers and altered molecular pathways. Am J Obstet Gynecol MFM 2024; 6:101535. [PMID: 39505208 DOI: 10.1016/j.ajogmf.2024.101535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/23/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Congenital diaphragmatic hernia (CDH) is characterized by a diaphragmatic defect, leading to herniation of abdominal organs into the chest, lung compression, and impaired lung development, often resulting in pulmonary hypertension and lung hypoplasia. Prenatal imaging techniques like ultrasound and MRI provide anatomical predictors of outcomes, but their limitations necessitate novel biomarkers for better prognostic accuracy. OBJECTIVE This study aims to identify unique circulating maternal, fetal, and neonatal microRNAs (miRNAs) that can distinguish CDH pregnancies from healthy controls and assess their potential as markers of disease severity. STUDY DESIGN We conducted a prospective study involving third-trimester maternal blood, amniotic fluid, cord blood, and neonatal blood samples from pregnancies complicated by CDH and healthy controls. miRNA expression was analyzed using RNA-sequencing, and random forest analysis identified miRNAs distinguishing CDH survivors from nonsurvivors. Pathway enrichment analyses were performed to explore the biological relevance of differentially expressed miRNAs. RESULTS Significant miRNA expression differences were observed between CDH and control samples across all sample types. In infant blood, 148 miRNAs were up-regulated, and 36 were down-regulated in CDH cases. Pathway analysis revealed that dysregulated miRNAs in CDH targeted pathways related to protein binding, transcription regulation, and signaling pathways implicated in pulmonary hypertension and lung hypoplasia. Random forest analysis identified miRNAs in maternal blood (miR-7850-5p_L-1R+2, miR-942-3p, and miR-197-3p) that distinguished CDH survivors from nonsurvivors, with an receiver operating characteristic area under the curve of 1.0. CONCLUSION Circulating miRNAs in maternal blood offer promising biomarkers for predicting CDH outcomes. miRNAs from infant blood provide mechanistic insights and potential targets for therapeutic intervention in critical pathways of pulmonary hypertension and lung hypoplasia. Further studies with larger cohorts are needed to validate these findings and explore the clinical application of miRNA biomarkers in CDH management.
Collapse
Affiliation(s)
- James R Bardill
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO (Bardill, Breckenfelder, Eason, Gallagher, and Derderian); Department of Surgery, Laboratory for Fetal and Regenerative Biology, University of Colorado Denver School of Medicine, Aurora, CO (Bardill, Breckenfelder, Eason, Khailova, and Derderian)
| | - Anis Karimpour-Fard
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO (Karimpour-Fard)
| | - Courtney C Breckenfelder
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO (Bardill, Breckenfelder, Eason, Gallagher, and Derderian); Department of Surgery, Laboratory for Fetal and Regenerative Biology, University of Colorado Denver School of Medicine, Aurora, CO (Bardill, Breckenfelder, Eason, Khailova, and Derderian)
| | - Carmen C Sucharov
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora, CO (Sucharov)
| | - Caitlin R Eason
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO (Bardill, Breckenfelder, Eason, Gallagher, and Derderian); Department of Surgery, Laboratory for Fetal and Regenerative Biology, University of Colorado Denver School of Medicine, Aurora, CO (Bardill, Breckenfelder, Eason, Khailova, and Derderian)
| | - Lauren T Gallagher
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO (Bardill, Breckenfelder, Eason, Gallagher, and Derderian)
| | - Ludmila Khailova
- Department of Surgery, Laboratory for Fetal and Regenerative Biology, University of Colorado Denver School of Medicine, Aurora, CO (Bardill, Breckenfelder, Eason, Khailova, and Derderian)
| | - Clyde J Wright
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO (Wright and Gien)
| | - Jason Gien
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO (Wright and Gien)
| | - Henry L Galan
- Colorado Fetal Care Center, Children's Hospital Colorado, Aurora, CO (Galan and Derderian); Divison of Maternal Fetal Medicine, University of Colorado School of Medicine, Aurora, CO (Galan)
| | - Sarkis Christopher Derderian
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO (Bardill, Breckenfelder, Eason, Gallagher, and Derderian); Department of Surgery, Laboratory for Fetal and Regenerative Biology, University of Colorado Denver School of Medicine, Aurora, CO (Bardill, Breckenfelder, Eason, Khailova, and Derderian); Colorado Fetal Care Center, Children's Hospital Colorado, Aurora, CO (Galan and Derderian); Division of Pediatric Surgery, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO (Derderian).
| |
Collapse
|
2
|
Liu C, Chen J, Huang X, Xia Q, Yang L, Guo J, Tian J, Wang J, Niu Y, Li L, Gou D. lncRNA VELRP Modulates Pulmonary Arterial Smooth Muscle Cell Proliferation and Promotes Vascular Remodeling in Pulmonary Hypertension. Arterioscler Thromb Vasc Biol 2024; 44:2560-2576. [PMID: 39360410 DOI: 10.1161/atvbaha.124.321416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Pulmonary hypertension is a devastating vascular disorder characterized by extensive pulmonary vascular remodeling, ultimately leading to right ventricular failure and death. Activation of PDGF (platelet-derived growth factor) signaling promotes the hyperproliferation of pulmonary arterial smooth muscle cells (PASMCs), thus contributing to the pulmonary vascular remodeling. However, the molecular mechanisms that govern hyperproliferation of PASMCs induced by PDGF remain largely unknown, including the contribution of long noncoding RNAs (lncRNAs). In this study, we aimed to identify a novel lncRNA regulated by PDGF implicated in PASMC proliferation in pulmonary vascular remodeling. METHODS RNA-sequencing analysis was conducted to identify a novel lncRNA named vessel-enriched lncRNA regulated by PDGF-BB (platelet-derived growth factor-BB; VELRP). Functional investigations of VELRP were performed using knockdown and overexpression strategies along with RNA sequencing. Validation of the function and potential mechanisms of VELRP was performed through Western blot, RNA immunoprecipitation, and chromatin immunoprecipitation assays. RESULTS We identified a novel vessel-enriched lncRNA with an increased response to PDGF-BB stimulus. VELRP was identified as an evolutionarily conserved RNA molecule. Modulation of VELRP in PASMCs significantly altered cell proliferation. Mechanistically, VELRP enhances trimethylation of H3K4 (histone H3 lysine 4) by interacting with WDR5 (WD repeat-containing protein 5), leading to increased expression of CDK (cyclin-dependent kinase) 1, CDK2, and CDK4 and consequent hyperproliferation of PASMCs. The pathological relevance of VELRP upregulation in pulmonary artery was confirmed using rat pulmonary hypertension models in vivo, as well as in PASMCs from patients with idiopathic pulmonary arterial hypertension. Specific knockdown of VELRP in smooth muscle cells using adeno-associated virus type 9 SM22α (smooth muscle protein 22α) promoter-shRNA-mediated silencing of VELRP resulted in a significant decrease in right ventricular systolic pressure and vascular remodeling in rat pulmonary hypertension model. CONCLUSIONS VELRP, as an lncRNA upregulated by PDGF-BB, mediates PASMC proliferation via WDR5/CDK signaling. In vivo studies demonstrate that targeted intervention of VELRP in smooth muscle cells can prevent the development of pulmonary hypertension.
Collapse
MESH Headings
- RNA, Long Noncoding/metabolism
- RNA, Long Noncoding/genetics
- Cell Proliferation
- Animals
- Vascular Remodeling
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiopathology
- Becaplermin/pharmacology
- Becaplermin/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/pathology
- Male
- Rats, Sprague-Dawley
- Cells, Cultured
- Disease Models, Animal
- Signal Transduction
- Rats
- Humans
- Histones/metabolism
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Arterial Hypertension/genetics
- Pulmonary Arterial Hypertension/pathology
Collapse
Affiliation(s)
- Cuilian Liu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Jidong Chen
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Xingtao Huang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Qinyi Xia
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Lei Yang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Jiao Guo
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Jinglin Tian
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Jun Wang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Yanqin Niu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Li Li
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| |
Collapse
|
3
|
Wei X, Li S, Yan H, Chen S, Li R, Zhang W, Chao S, Guo W, Li W, Ahmed Z, Lei C, Ma Z. Unraveling genomic diversity and positive selection signatures of Qaidam cattle through whole-genome re-sequencing. Anim Genet 2024; 55:362-376. [PMID: 38480515 DOI: 10.1111/age.13417] [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: 01/02/2024] [Revised: 01/02/2024] [Accepted: 02/22/2024] [Indexed: 05/04/2024]
Abstract
Qaidam cattle are a typical Chinese native breed inhabiting northwest China. They bear the characteristics of high cold and roughage tolerance, low-oxygen adaptability and good meat quality. To analyze the genetic diversity of Qaidam cattle, 60 samples were sequenced using whole-genome resequencing technology, along with 192 published sets of whole-genome sequencing data of Indian indicine cattle, Chinese indicine cattle, North Chinese cattle breeds, East Asian taurine cattle, Eurasian taurine cattle and European taurine cattle as controls. It was found that Qaidam cattle have rich genetic diversity in Bos taurus, but the degree of inbreeding is also high, which needs further protection. The phylogenetic analysis, principal component analysis and ancestral component analysis showed that Qaidam cattle mainly originated from East Asian taurine cattle. Qaidam cattle had a closer genetic relationship with the North Chinese cattle breeds and the least differentiation from Mongolian cattle. Annotating the selection signals obtained by composite likelihood ratio, nucleotide diversity analysis, integrated haplotype score, genetic differentiation index, genetic diversity ratio and cross-population extended haplotype homozygosity methods, several genes associated with immunity, reproduction, meat, milk, growth and adaptation showed strong selection signals. In general, this study provides genetic evidence for understanding the germplasm characteristics of Qaidam cattle. At the same time, it lays a foundation for the scientific and reasonable protection and utilization of genetic resources of Chinese local cattle breeds, which has great theoretical and practical significance.
Collapse
Affiliation(s)
- Xudong Wei
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Xining, China
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Xining, China
| | - Shuang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Huixuan Yan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Shengmei Chen
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Xining, China
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Xining, China
| | - Ruizhe Li
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Xining, China
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Xining, China
| | - Weizhong Zhang
- Golmud Animal Husbandry and Veterinary Station of Qinghai Province, Golmud, China
| | - Shengyu Chao
- Agro-Technical Extension and Service Center in Haixi Prefecture of Qinghai Province, Delingha, China
| | - Weixing Guo
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Xining, China
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Xining, China
| | - Wenhao Li
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Xining, China
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Xining, China
| | - Zulfiqar Ahmed
- Department of Livestock and Poultry Production, Faculty of Veterinary and Animal Sciences, University of Poonch Rawalakot, Rawalakot, Pakistan
| | - Chuzhao Lei
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhijie Ma
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Xining, China
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Xining, China
| |
Collapse
|
4
|
Hang C, Zu L, Luo X, Wang Y, Yan L, Zhang Z, Le K, Huang Y, Ye L, Ying Y, Chen K, Xu X, Lv Q, Du L. Ddx5 Targeted Epigenetic Modification of Pericytes in Pulmonary Hypertension After Intrauterine Growth Restriction. Am J Respir Cell Mol Biol 2024; 70:400-413. [PMID: 38301267 DOI: 10.1165/rcmb.2023-0244oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 02/01/2024] [Indexed: 02/03/2024] Open
Abstract
Newborns with intrauterine growth restriction (IUGR) have a higher likelihood of developing pulmonary arterial hypertension (PAH) in adulthood. Although there is increasing evidence suggesting that pericytes play a role in regulating myofibroblast transdifferentiation and angiogenesis in malignant and cardiovascular diseases, their involvement in the pathogenesis of IUGR-related pulmonary hypertension and the underlying mechanisms remain incompletely understood. To address this issue, a study was conducted using a Sprague-Dawley rat model of IUGR-related pulmonary hypertension. Our investigation revealed increased proliferation and migration of pulmonary microvascular pericytes in IUGR-related pulmonary hypertension, accompanied by weakened endothelial-pericyte interactions. Through whole-transcriptome sequencing, Ddx5 (DEAD-box protein 5) was identified as one of the hub genes in pericytes. DDX5, a member of the RNA helicase family, plays a role in the regulation of ATP-dependent RNA helicase activities and cellular function. MicroRNAs have been implicated in the pathogenesis of PAH, and microRNA-205 (miR-205) regulates cell proliferation, migration, and angiogenesis. The results of dual-luciferase reporter assays confirmed the specific binding of miR-205 to Ddx5. Mechanistically, miR-205 negatively regulates Ddx5, leading to the degradation of β-catenin by inhibiting the phosphorylation of Gsk3β at serine 9. In vitro experiments showed the addition of miR-205 effectively ameliorated pericyte dysfunction. Furthermore, in vivo experiments demonstrated that miR-205 agomir could ameliorate pulmonary hypertension. Our findings indicated that the downregulation of miR-205 expression mediates pericyte dysfunction through the activation of Ddx5. Therefore, targeting the miR-205/Ddx5/p-Gsk3β/β-catenin axis could be a promising therapeutic approach for IUGR-related pulmonary hypertension.
Collapse
Affiliation(s)
| | - Lu Zu
- Department of Neonatology and
| | - Xiaofei Luo
- Department of Pediatrics, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China; and
| | - Yu Wang
- Department of Neonatology and
| | - Lingling Yan
- Department of Pediatrics, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China; and
| | | | - Kaixing Le
- Academy of Pediatrics, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | | | | | | | | | - Xuefeng Xu
- Department of Rheumatology, Immunology, and Allergy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, People's Republic of China
| | | | | |
Collapse
|
5
|
Zervou MI, Tarlatzis BC, Grimbizis GF, Spandidos DA, Niewold TB, Goulielmos GN. Association of endometriosis with Sjögren's syndrome: Genetic insights (Review). Int J Mol Med 2024; 53:20. [PMID: 38186322 PMCID: PMC10781419 DOI: 10.3892/ijmm.2024.5344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 12/20/2023] [Indexed: 01/09/2024] Open
Abstract
Patients with a history of endometriosis have an increased risk of developing various autoimmune diseases such as rheumatoid arthritis, ankylosing spondylitis, systemic lupus erythematosus, multiple sclerosis and celiac disease. There is a potential association between endometriosis and an increased susceptibility for Sjögren's syndrome (SS). SS is a common chronic, inflammatory, systemic, autoimmune, multifactorial disease of complex pathology, with genetic, epigenetic and environmental factors contributing to the development of this condition. It occurs in 0.5‑1% of the population, is characterized by the presence of ocular dryness, lymphocytic infiltrations and contributes to neurological, gastrointestinal, vascular and dermatological manifestations. Endometriosis is an inflammatory, estrogen‑dependent, multifactorial, heterogeneous gynecological disease, affecting ≤10% of reproductive‑age women. It is characterized by the occurrence of endometrial tissue outside the uterine cavity, mainly in the pelvic cavity, and is associated with pelvic pain, dysmenorrhea, deep dyspareunia and either subfertility or infertility. It is still unclear whether SS appears as a secondary response to endometriosis, or it is developed due to any potential shared mechanisms of these conditions. The aim of the present review was to explore further the biological basis only of the co‑occurrence of these disorders but not their association at clinical basis, focusing on the analysis of the partially shared genetic background between endometriosis and SS, and the clarification of the possible similarities in the underlying pathogenetic mechanisms and the relevant molecular pathways.
Collapse
Affiliation(s)
- Maria I. Zervou
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, 71403 Heraklion, Greece
| | - Basil C. Tarlatzis
- First Department of Obstetrics and Gynecology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Grigoris F. Grimbizis
- Unit for Human Reproduction, First Department of Obstetrics and Gynecology, 'Papageorgiou' General Hospital, Aristotle University Medical School, 56403 Thessaloniki, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71403 Heraklion, Greece
| | - Timothy B. Niewold
- Barbara Volcker Center for Women and Rheumatic Disease, New York, NY 10021, USA
- Hospital for Special Surgery, New York, NY 10021, USA
| | - George N. Goulielmos
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, 71403 Heraklion, Greece
- Department of Internal Medicine, University Hospital of Heraklion, 71500 Heraklion, Greece
| |
Collapse
|
6
|
Cho S, Oh SB, Kim HJ, Kim SJ. T18/S19 diphosphorylation of myosin regulatory light chain impairs pulmonary artery relaxation in monocrotaline-induced pulmonary hypertensive rats. Pflugers Arch 2023; 475:1097-1112. [PMID: 37422604 DOI: 10.1007/s00424-023-02836-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/29/2023] [Accepted: 06/26/2023] [Indexed: 07/10/2023]
Abstract
Phosphorylation of Ser19 (S19-p) on the myosin regulatory light chain (MLC2) is critical for arterial contraction. It has been shown that elevated RhoA-dependent kinase (ROCK) activity or decreased MLC phosphatase (MLCP) activity leads to further phosphorylation of Thr18 (T18/S19-pp), which has been linked to vasospastic diseases. However, this phenomenon has not yet been studied in the context of pulmonary arterial hypertension (PAH). In the monocrotaline-induced PAH (PAH-MCT) rat model, we observed a significant delay in pulmonary artery (PA) relaxation following high potassium-induced contraction, which persisted even with the use of an L-type calcium channel blocker or in a calcium-free solution. Immunoblot analysis showed increased levels of both S19-p and T18/S19-pp in unstimulated PAs from PAH-MCT rats. Proteomics analysis revealed a reduction in soluble guanylate cyclase (sGC) and protein kinase G (PKG) levels, and immunoblotting confirmed decreased levels of MYPT1 (a component of MLCP) and increased ROCK in PAH-MCT. In the control PAs, the pharmacological inhibition of sGC with ODQ resulted in a prominent delay of relaxation and increased T18/S19-pp as in PAH-MCT. The delayed relaxation and the T18/S19-pp in PAH-MCT were reversed by ROCK inhibitor, Y27632, while not by membrane permeable 8-Br-cGMP. The delayed relaxation and T18/S19-diP in the ODQ-treated control PA were also reversed by Y27632. Taken together, the decreased sGC and MLCP, and increased ROCK increased T18/S19-pp, which leads to the decreased ability of PA to relax in PAH-MCT rats. PA specific inhibition of ROCK or activation of MLCP are expected to serve as potential drugs in the treatment of PAH.
Collapse
Affiliation(s)
- Suhan Cho
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Seung Beom Oh
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Hae Jin Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, MO, Columbia, USA
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
7
|
Zaoui K, Duhamel S. RhoB as a tumor suppressor: It’s all about localization. Eur J Cell Biol 2023; 102:151313. [PMID: 36996579 DOI: 10.1016/j.ejcb.2023.151313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/15/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
The small GTPase RhoB is distinguished from other Rho proteins by its unique subcellular localization in endosomes, multivesicular bodies, and nucleus. Despite high sequence homology with RhoA and RhoC, RhoB is mainly associated with tumor suppressive function, while RhoA and RhoC support oncogenic transformation in most malignancies. RhoB regulates the endocytic trafficking of signaling molecules and cytoskeleton remodeling, thereby controlling growth, apoptosis, stress response, immune function, and cell motility in various contexts. Some of these functions may be ascribed to RhoB's unique subcellular localization to endocytic compartments. Here we describe the pleiotropic roles of RhoB in cancer suppression in the context of its subcellular localization, and we discuss possible therapeutic avenues to pursue and highlight priorities for future research.
Collapse
|
8
|
Pugnaloni F, Capolupo I, Patel N, Giliberti P, Dotta A, Bagolan P, Kipfmueller F. Role of microRNAs in Congenital Diaphragmatic Hernia-Associated Pulmonary Hypertension. Int J Mol Sci 2023; 24:ijms24076656. [PMID: 37047629 PMCID: PMC10095389 DOI: 10.3390/ijms24076656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Epigenetic regulators such as microRNAs (miRNAs) have a key role in modulating several gene expression pathways and have a role both in lung development and function. One of the main pathogenetic determinants in patients with congenital diaphragmatic hernia (CDH) is pulmonary hypertension (PH), which is directly related to smaller lung size and pulmonary microarchitecture alterations. The aim of this review is to highlight the importance of miRNAs in CDH-related PH and to summarize the results covering this topic in animal and human CDH studies. The focus on epigenetic modulators of CDH-PH offers the opportunity to develop innovative diagnostic tools and novel treatment modalities, and provides a great potential to increase researchers’ understanding of the pathophysiology of CDH.
Collapse
Affiliation(s)
- Flaminia Pugnaloni
- Neonatal Intensive Care Unit, Bambino Gesù Children Hospital, Instituti di Ricovero e Cura a Carattere Scietifico (IRCCS), 00165 Rome, Italy
| | - Irma Capolupo
- Neonatal Intensive Care Unit, Bambino Gesù Children Hospital, Instituti di Ricovero e Cura a Carattere Scietifico (IRCCS), 00165 Rome, Italy
| | - Neil Patel
- Department of Neonatology, The Royal Hospital for Children, Glasgow G51 4TF, UK
| | - Paola Giliberti
- Neonatal Intensive Care Unit, Bambino Gesù Children Hospital, Instituti di Ricovero e Cura a Carattere Scietifico (IRCCS), 00165 Rome, Italy
| | - Andrea Dotta
- Neonatal Intensive Care Unit, Bambino Gesù Children Hospital, Instituti di Ricovero e Cura a Carattere Scietifico (IRCCS), 00165 Rome, Italy
| | - Pietro Bagolan
- Area of Fetal, Neonatal and Cardiological Sciences Children’s Hospital Bambino Gesù-Research Institute, 00165 Rome, Italy
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00165 Rome, Italy
| | - Florian Kipfmueller
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
9
|
Zhao M, Liu J, Xin M, Yang K, Huang H, Zhang W, Zhang J, He S. Pulmonary arterial hypertension associated with congenital heart disease: An omics study. Front Cardiovasc Med 2023; 10:1037357. [PMID: 36970344 PMCID: PMC10036813 DOI: 10.3389/fcvm.2023.1037357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/24/2023] [Indexed: 03/12/2023] Open
Abstract
Pulmonary arterial hypertension associated with congenital heart disease (PAH-CHD) is a severely progressive condition with uncertain physiological course. Hence, it has become increasingly relevant to clarify the specific mechanisms of molecular modification, which is crucial to identify more treatment strategies. With the rapid development of high-throughput sequencing, omics technology gives access to massive experimental data and advanced techniques for systems biology, permitting comprehensive assessment of disease occurrence and progression. In recent years, significant progress has been made in the study of PAH-CHD and omics. To provide a comprehensive description and promote further in-depth investigation of PAH-CHD, this review attempts to summarize the latest developments in genomics, transcriptomics, epigenomics, proteomics, metabolomics, and multi-omics integration.
Collapse
Affiliation(s)
- Maolin Zhao
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Jian Liu
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Mei Xin
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Ke Yang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Honghao Huang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Wenxin Zhang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Jinbao Zhang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Siyi He
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
- Correspondence: Siyi He
| |
Collapse
|
10
|
Discovering novel clues of natural selection on four worldwide goat breeds. Sci Rep 2023; 13:2110. [PMID: 36747064 PMCID: PMC9902602 DOI: 10.1038/s41598-023-27490-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/03/2023] [Indexed: 02/08/2023] Open
Abstract
In goat breeds, the domestication followed by artificial selection for economically important traits have shaped genetic variation within populations, leading to the fixation of specific alleles for specific traits. This led to the formation and evolution of many different breeds specialised and raised for a particular purpose. However, and despite the intensity of artificial selection, natural selection continues acting, possibly leaving a more diluted contribution over time, whose traces may be more difficult to capture. In order to explore selection footprints as response of environmental adaptation, we analysed a total of 993 goats from four transboundary goats breeds (Angora, Boer, Nubian and Saanen) genotyped with the SNP chip 50 K using outlier detection, runs of homozygosity and haplotype-based detection methods. Our results showed that all methods identified footprints on chromosome 6 (from 30 to 49 Mb) for two specific populations of Nubian goats sampled in Egypt. In Angora and Saanen breeds, we detected two selective sweeps using HapFLK, on chromosome 21 (from 52 to 55 Mb) and chromosome 25 (from 1 to 5 Mb) respectively. The analysis of runs of homozygosity showed some hotspots in all breeds. The overall investigation of the selected regions detected combining the different approaches and the gene ontology exploration revealed both novel and well-known loci related to adaptation, especially for heat stress. Our findings can help to better understand the balance between the two selective pressures in commercial goat breeds providing new insights on the molecular mechanisms of adaptation.
Collapse
|
11
|
Effect of positive pressure ventilation and bariatric surgery on extracellular vesicle microRNAs in patients with severe obesity and obstructive sleep apnea. Int J Obes (Lond) 2023; 47:24-32. [PMID: 36284205 DOI: 10.1038/s41366-022-01230-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Obstructive sleep apnea (OSA) and severe obesity share a common pathophysiological phenomenon, systemic and tissue hypoxia. Hypoxaemia modifies microRNA expression, particularly, extracellular vesicles microRNAs which are involved in the progression of cardiovascular diseases, metabolic syndrome and cancer. We aim to evaluate extracellular vesicle miRNAs among patients with severe obesity with and without OSA and the effect of OSA and severe obesity treatment: continuous positive airway pressure (CPAP) and bariatric surgery. METHODS Patients were selected from the Epigenetics Modification in Morbid Obesity and Obstructive Sleep Apnea (EPIMOOSA) study (NCT03995836), a prospective observational study of patients undergoing bariatric surgery. Patients were divided into OSA (Apnea-hyponea index (AHI) > 10) and non-OSA (AHI < 10). Patients with OSA were treated with CPAP for 6 months. Then, all patients had bariatric surgery and re-evaluated 12 months later. At each visit, blood samples were obtained for biobanking. Subsequently, extracellular vesicles were extracted, and then, miRNA expression was analysed. RESULTS 15 patients with OSA and 9 without OSA completed the protocol. At baseline, patients with OSA showed higher miR16, miR126 and miR320 (p < 0.05) and lower miR223 expression (p < 0.05) than those without OSA. In patients with severe obesity and OSA, after 6 months with CPAP, we observed a significant decrease in miR21 (p < 0.01), miR126 (p < 0.001) and miR320 (p < 0.001), with no changes in any miRNA in patients without OSA. No changes were detected in any miRNA after 6 months of bariatric surgery in patients with or without OSA. CONCLUSION Co-existance of OSA and severe obesity alters the profile of extracellular vesicle miRNAs. Bariatric surgery and weight loss did not reverse this effect meanwhile the treatment with CPAP in patients with severe obesity and OSA showed a recovery outcome in those extracellular vesicle miRNAs. Those facts remark the need for OSA screening in patients with severe obesity. CLINICAL TRIAL REGISTRATION The study has also been registered at ClinicalTrials.gov identifier: NCT03995836.
Collapse
|
12
|
Mechanism of Hypoxia-Mediated Smooth Muscle Cell Proliferation Leading to Vascular Remodeling. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3959845. [PMID: 36593773 PMCID: PMC9805398 DOI: 10.1155/2022/3959845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/25/2022] [Accepted: 12/07/2022] [Indexed: 12/25/2022]
Abstract
Vascular remodeling refers to changes in the size, contraction, distribution, and flow rate of blood vessels and even changes in vascular function. Vascular remodeling can cause cardiovascular and cerebrovascular diseases. It can also lead to other systemic diseases, such as pulmonary hypertension, pulmonary atherosclerosis, chronic obstructive pulmonary disease, stroke, and ascites of broilers. Hypoxia is one of the main causes of vascular remodeling. Prolonged hypoxia or intermittent hypoxia can lead to loss of lung ventilation, causing respiratory depression, irregular respiratory rhythms, and central respiratory failure. Animals that are unable to adapt to the highland environment are also prone to sustained constriction of the small pulmonary arteries, increased resistance to pulmonary circulation, and impaired blood circulation, leading to pulmonary hypertension and right heart failure if they live in a highland environment for long periods of time. However, limited studies have been found on the relationship between hypoxia and vascular remodeling. Therefore, this review will explore the relationship between hypoxia and vascular remodeling from the aspects of endoplasmic reticulum stress, mitochondrial dysfunction, abnormal calcium channel, disordered cellular metabolism, abnormal expression of miRNA, and other factors. This will help to understand the detailed mechanism of hypoxia-mediated smooth muscle cell proliferation and vascular remodeling for the better treatment and management of diseases due to vascular remodeling.
Collapse
|
13
|
Yang M, Liao M, Liu R, Zhang Q, Zhang S, He Y, Jin J, Zhang P, Zhou L. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles loaded with miR-223 ameliorate myocardial infarction through P53/S100A9 axis. Genomics 2022; 114:110319. [PMID: 35227836 DOI: 10.1016/j.ygeno.2022.110319] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/22/2022] [Accepted: 02/19/2022] [Indexed: 01/14/2023]
Abstract
Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have been proposed as a promising strategy for myocardial infarction (MI). This study aims to explore the mechanism of human umbilical cord MSCs (hucMSCs)-derived EVs loaded with miR-223 on MI. Inflammation, cell biological functions, and fibrosis in vitro were measured. Furthermore, MI rat models were established to verify the role of EVs-miR-223 in vivo. The binding relationship between miR-223 and P53 was confirmed. ChIP assay was utilized to observe the combination of P53 and S100A9. The suppressed fibrosis of cardiomyocytes occurred with cells overexpressing miR-223. MiR-223 contributed to the angiogenesis of HUVECs. P53 was a target gene of miR-223. In vivo, miR-223 relieved myocardial fibrosis and inflammation infiltration, and promoted the angiogenesis in MI rats. HucMSC-derived EVs loaded with miR-223 mitigates MI and promotes myocardial repair through the P53/S100A9 axis, manifesting the underlying therapy values of hucMSC-derived EVs loaded with miR-223 in MI.
Collapse
Affiliation(s)
- Mei Yang
- Departmemt of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Mingmei Liao
- Departmemt of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Ruijie Liu
- Departmemt of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Qi Zhang
- Departmemt of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Sai Zhang
- NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yi He
- NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Jin Jin
- NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Pengfei Zhang
- NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China.
| | - Lin Zhou
- Departmemt of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
14
|
MicroRNAs in Pulmonary Hypertension, from Pathogenesis to Diagnosis and Treatment. Biomolecules 2022; 12:biom12040496. [PMID: 35454085 PMCID: PMC9031307 DOI: 10.3390/biom12040496] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/12/2022] [Accepted: 03/14/2022] [Indexed: 02/04/2023] Open
Abstract
Pulmonary hypertension (PH) is a fatal and untreatable disease, ultimately leading to right heart failure and eventually death. microRNAs are small, non-coding endogenous RNA molecules that can regulate gene expression and influence various biological processes. Changes in microRNA expression levels contribute to various cardiovascular disorders, and microRNAs have been shown to play a critical role in PH pathogenesis. In recent years, numerous studies have explored the role of microRNAs in PH, focusing on the expression profiles of microRNAs and their signaling pathways in pulmonary artery smooth muscle cells (PASMCs) or pulmonary artery endothelial cells (PAECs), PH models, and PH patients. Moreover, certain microRNAs, such as miR-150 and miR-26a, have been identified as good candidates of diagnosis biomarkers for PH. However, there are still several challenges for microRNAs as biomarkers, including difficulty in normalization, specificity in PH, and a lack of longitudinal and big sample-sized studies. Furthermore, microRNA target drugs are potential therapeutic agents for PH treatment, which have been demonstrated in PH models and in humans. Nonetheless, synthetic microRNA mimics or antagonists are susceptible to several common defects, such as low drug efficacy, inefficient drug delivery, potential toxicity and especially, off-target effects. Therefore, finding clinically safe and effective microRNA drugs remains a great challenge, and further breakthrough is urgently needed.
Collapse
|
15
|
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
|
16
|
Deng Y, Tong J, Shi W, Tian Z, Yu B, Tang J. Thromboangiitis obliterans plasma-derived exosomal miR-223-5p inhibits cell viability and promotes cell apoptosis of human vascular smooth muscle cells by targeting VCAM1. Ann Med 2021; 53:1129-1141. [PMID: 34259105 PMCID: PMC8281010 DOI: 10.1080/07853890.2021.1949487] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/24/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Exosomes-encapsulated microRNAs (miRNAs) have been established to be implicated in the pathogenesis of different diseases. Nevertheless, circulating exosomal miRNAs of thromboangiitis obliterans (TAO) remains poorly understood. This study aimed to explore the effects of exosomal miRNAs associated with TAO on human vascular smooth muscle cells (HVSMCs).Methods: The exosomes were isolated from the plasma of TAO patients and normal controls and then were sent for small RNA sequencing. Differentially expressed miRNAs (DE-miRNAs) were identified by bioinformatics analysis and were confirmed by RT-qPCR. After that, PKH67 staining was used to label exosomes and co-cultured with HVSMCs. Cell viability and apoptosis were, respectively, tested by CCK-8 assay and flow cytometry. Finally, dual-luciferase reporter assay was used to confirm the downstream targets of miR-223-5p.Results: A total of 39 DE-miRNAs were identified between TAO patients and normal controls, of which, miR-223-5p was one of the most significantly up-regulated miRNAs. TAO plasma-derived exosomes or miR-223-5p mimics inhibited cell viability of HVSMCs and promoted cell apoptosis. The pro-apoptotic effect of TAO plasma-derived exosomes was alleviated by miR-223-5p inhibitor. Additionally, the expressions of VCAM1 and IGF1R were down-regulated by exosomes and miR-223-5p mimics, and were abrogated by miR-223-5p inhibitor. Dual-luciferase report showed that VCAM1 was the target of miR-223-5p.Conclusions: Our findings imply that circulating exosomal miR-223-5p may play an essential role in the pathogenesis of TAO, and provide a basis for miR-6515-5p/VCAM1 as novel therapeutic targets and pathways for TAO treatment.
Collapse
Affiliation(s)
- Ying Deng
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jindong Tong
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| | - Weijun Shi
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zhongyi Tian
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| | - Bo Yu
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jingdong Tang
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
17
|
Extracellular Vesicles and Their miRNA Content in Amniotic and Tracheal Fluids of Fetuses with Severe Congenital Diaphragmatic Hernia Undergoing Fetal Intervention. Cells 2021; 10:cells10061493. [PMID: 34198576 PMCID: PMC8231823 DOI: 10.3390/cells10061493] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/18/2022] Open
Abstract
Infants with congenital diaphragmatic hernia (CDH) are at high risk of postnatal mortality due to lung hypoplasia and arterial pulmonary hypertension. In severe cases, prenatal intervention by fetal endoscopic tracheal occlusion (FETO) can improve survival by accelerating lung growth. However, postnatal mortality remains in the range of about 50% despite fetal treatment, and there is currently no clear explanation for this different clinical response to FETO. We evaluated the concentration of extracellular vesicles (EVs) and associated microRNA expression in amniotic and tracheal fluids of fetuses with CDH undergoing FETO, and we examined the association between molecular findings and postnatal survival. We observed a higher count of EVs in the amniotic fluid of non-survivors and in the tracheal fluid sampled in utero at the time of reversal of tracheal occlusion, suggesting a pro-inflammatory lung reactivity that is already established in utero and that could be associated with a worse postnatal clinical course. In addition, we observed differential regulation of four EV-enclosed miRNAs (miR-379-5p, miR-889-3p; miR-223-3p; miR-503-5p) in relation to postnatal survival, with target genes possibly involved in altered lung development. Future research should investigate molecular therapeutic agents targeting differentially regulated miRNAs to normalize their expression and potentially improve clinical outcomes.
Collapse
|
18
|
Jiao P, Wang XP, Luoreng ZM, Yang J, Jia L, Ma Y, Wei DW. miR-223: An Effective Regulator of Immune Cell Differentiation and Inflammation. Int J Biol Sci 2021; 17:2308-2322. [PMID: 34239357 PMCID: PMC8241730 DOI: 10.7150/ijbs.59876] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/21/2021] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs) play a critical role in regulating various biological processes, such as cell differentiation and immune modulation by binding to their target genes. miR-223 is a miRNA with important functions and has been widely investigated in recent years. Under certain physiological conditions, miR-223 is regulated by different transcription factors, including sirtuin1 (Sirt1), PU.1 and Mef2c, and its biological functions are mediated through changes in its cellular or tissue expression. This review paper summarizes miR-223 biosynthesis and its regulatory role in the differentiation of granulocytes, dendritic cells (DCs) and lymphocytes, macrophage polarization, and endothelial and epithelial inflammation. In addition, it describes the molecular mechanisms of miR-223 in regulating lung inflammation, rheumatoid arthritis, enteritis, neuroinflammation and mastitis to provide insights into the existing molecular regulatory networks and therapies for inflammatory diseases in humans and animals.
Collapse
Affiliation(s)
- Peng Jiao
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Xing-Ping Wang
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Zhuo-Ma Luoreng
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Jian Yang
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Li Jia
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Yun Ma
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Da-Wei Wei
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| |
Collapse
|
19
|
Kostyunina DS, McLoughlin P. Sex Dimorphism in Pulmonary Hypertension: The Role of the Sex Chromosomes. Antioxidants (Basel) 2021; 10:779. [PMID: 34068984 PMCID: PMC8156365 DOI: 10.3390/antiox10050779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 01/01/2023] Open
Abstract
Pulmonary hypertension (PH) is a condition characterised by an abnormal elevation of pulmonary artery pressure caused by an increased pulmonary vascular resistance, frequently leading to right ventricular failure and reduced survival. Marked sexual dimorphism is observed in patients with pulmonary arterial hypertension, a form of pulmonary hypertension with a particularly severe clinical course. The incidence in females is 2-4 times greater than in males, although the disease is less severe in females. We review the contribution of the sex chromosomes to this sex dimorphism highlighting the impact of proteins, microRNAs and long non-coding RNAs encoded on the X and Y chromosomes. These genes are centrally involved in the cellular pathways that cause increased pulmonary vascular resistance including the production of reactive oxygen species, altered metabolism, apoptosis, inflammation, vasoconstriction and vascular remodelling. The interaction with genetic mutations on autosomal genes that cause heritable pulmonary arterial hypertension such as bone morphogenetic protein 2 (BMPR2) are examined. The mechanisms that can lead to differences in the expression of genes located on the X chromosomes between females and males are also reviewed. A better understanding of the mechanisms of sex dimorphism in this disease will contribute to the development of more effective therapies for both women and men.
Collapse
Affiliation(s)
| | - Paul McLoughlin
- Conway Institute, School of Medicine, University College Dublin, Dublin D04 V1W8, Ireland;
| |
Collapse
|
20
|
Zhang MW, Shen YJ, Shi J, Yu JG. MiR-223-3p in Cardiovascular Diseases: A Biomarker and Potential Therapeutic Target. Front Cardiovasc Med 2021; 7:610561. [PMID: 33553260 PMCID: PMC7854547 DOI: 10.3389/fcvm.2020.610561] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/23/2020] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases, involving vasculopathy, cardiac dysfunction, or circulatory disturbance, have become the major cause of death globally and brought heavy social burdens. The complexity and diversity of the pathogenic factors add difficulties to diagnosis and treatment, as well as lead to poor prognosis of these diseases. MicroRNAs are short non-coding RNAs to modulate gene expression through directly binding to the 3′-untranslated regions of mRNAs of target genes and thereby to downregulate the protein levels post-transcriptionally. The multiple regulatory effects of microRNAs have been investigated extensively in cardiovascular diseases. MiR-223-3p, expressed in multiple cells such as macrophages, platelets, hepatocytes, and cardiomyocytes to modulate their cellular activities through targeting a variety of genes, is involved in the pathological progression of many cardiovascular diseases. It participates in regulation of several crucial signaling pathways such as phosphatidylinositol 3-kinase/protein kinase B, insulin-like growth factor 1, nuclear factor kappa B, mitogen-activated protein kinase, NOD-like receptor family pyrin domain containing 3 inflammasome, and ribosomal protein S6 kinase B1/hypoxia inducible factor 1 α pathways to affect cell proliferation, migration, apoptosis, hypertrophy, and polarization, as well as electrophysiology, resulting in dysfunction of cardiovascular system. Here, in this review, we will discuss the role of miR-223-3p in cardiovascular diseases, involving its verified targets, influenced signaling pathways, and regulation of cell function. In addition, the potential of miR-223-3p as therapeutic target and biomarker for diagnosis and prediction of cardiovascular diseases will be further discussed, providing clues for clinicians.
Collapse
Affiliation(s)
- Meng-Wan Zhang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yun-Jie Shen
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Shi
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Guang Yu
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
21
|
Su F, Shi M, Zhang J, Zheng Q, Wang H, Li X, Chen J. MiR-223/NFAT5 signaling suppresses arterial smooth muscle cell proliferation and motility in vitro. Aging (Albany NY) 2020; 12:26188-26198. [PMID: 33373321 PMCID: PMC7803580 DOI: 10.18632/aging.202395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 10/31/2020] [Indexed: 01/11/2023]
Abstract
Aberrant proliferation and migration of vascular smooth muscle cells contributes to cardiovascular diseases (CVDs), including atherosclerosis. MicroRNA-223 (miR-223) protects against atherosclerotic CVDs. We investigated the contribution of miR-223 to platelet-derived growth factor-BB (PDGF-BB)-induced proliferation and migration of human aortic smooth muscle cells (HASMCs). We found that miR-223 was downregulated in PDGF-BB-treated HASMCs in a dose- and time-dependent manner, while nuclear factor of activated T cells 5 (NFAT5) was upregulated. Gain- and loss-of-function studies demonstrated that miR-223 treatment reduced PDGF-BB-induced HASMC proliferation and motility, whereas miR-223 inhibitor enhanced these processes. Moreover, NFAT5 was identified as a direct target of miR-223 in HASMC. The inhibitory effects of miR-223 on HASMC proliferation and migration were partly rescued by NFAT5 restoration. Overall, these findings suggest that miR-223 inhibits the PDGF-BB-induced proliferation and motility of HASMCs by targeting NFAT5 and that miR-223 and NFAT5 may be potential therapeutic targets for atherosclerosis.
Collapse
Affiliation(s)
- Feifei Su
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China.,Department of Cardiology, Air Force General Hospital, PLA, Beijing 100142, China
| | - Miaoqian Shi
- Department of Cardiology, PLA Army General Hospital, Beijing 100700, China
| | - Jian Zhang
- Department of Cardiology, Beijing Chest Hospital Heart Center, Capital Medical University, Beijing 101149, China
| | - Qiangsun Zheng
- Division of Cardiology, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi'an 710004, Shaanxi, China
| | - Haichang Wang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China
| | - Xue Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China
| | - Jianghong Chen
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China
| |
Collapse
|
22
|
Guo J, Zhang L, Lian L, Hao M, Chen S, Hong Y. CircATP2B4 promotes hypoxia-induced proliferation and migration of pulmonary arterial smooth muscle cells via the miR-223/ATR axis. Life Sci 2020; 262:118420. [DOI: 10.1016/j.lfs.2020.118420] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022]
|
23
|
Santos-Ferreira CA, Abreu MT, Marques CI, Gonçalves LM, Baptista R, Girão HM. Micro-RNA Analysis in Pulmonary Arterial Hypertension: Current Knowledge and Challenges. ACTA ACUST UNITED AC 2020; 5:1149-1162. [PMID: 33294743 PMCID: PMC7691282 DOI: 10.1016/j.jacbts.2020.07.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 01/18/2023]
Abstract
The role of miRNAs in PAH is fast expanding, and it is increasingly difficult to identify which molecules have the highest translational potential. This review discusses the challenges in miRNA analysis and interpretation in PAH and highlights 4 promising miRNAs in this field. Additional pre-clinical studies and clinical trials are urgently needed to bring miRNAs from the bench to the bedside soon.
Pulmonary arterial hypertension (PAH) is a rare, chronic disease of the pulmonary vasculature that is associated with poor outcomes. Its pathogenesis is multifactorial and includes micro-RNA (miRNA) deregulation. The understanding of the role of miRNAs in PAH is expanding quickly, and it is increasingly difficult to identify which miRNAs have the highest translational potential. This review summarizes the current knowledge of miRNA expression in PAH, discusses the challenges in miRNA analysis and interpretation, and highlights 4 promising miRNAs in this field (miR-29, miR-124, miR-140, and miR-204).
Collapse
Key Words
- BMPR2, bone morphogenetic protein receptor type 2
- EPC, endothelial progenitor cell
- HIF, hypoxia-inducible factor
- HPAH, hereditary pulmonary arterial hypertension
- MCT, monocrotaline
- PAAF, pulmonary arterial adventitial fibroblast
- PAEC, pulmonary artery endothelial cell
- PAH, pulmonary arterial hypertension
- PASMC, pulmonary artery smooth muscle cells
- PH, pulmonary hypertension
- RV, right ventricle
- SU/Hx/Nx, association of Sugen 5416 with chronic hypoxia followed by normoxia
- WHO, World Health Organization
- animal model
- lncRNA, long noncoding RNA
- mRNA, messenger RNA
- miRNA, micro-RNA
- micro-RNA
- microarray
- ncRNAs, noncoding RNAs
- pulmonary arterial hypertension
Collapse
Affiliation(s)
- Cátia A Santos-Ferreira
- Cardiology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | - Mónica T Abreu
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | - Carla I Marques
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | - Lino M Gonçalves
- Cardiology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | - Rui Baptista
- Cardiology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal.,Cardiology Department, Centro Hospitalar Entre Douro e Vouga, Santa Maria de Feira, Portugal
| | - Henrique M Girão
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal
| |
Collapse
|
24
|
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
|
25
|
Ghoreishifar SM, Eriksson S, Johansson AM, Khansefid M, Moghaddaszadeh-Ahrabi S, Parna N, Davoudi P, Javanmard A. Signatures of selection reveal candidate genes involved in economic traits and cold acclimation in five Swedish cattle breeds. Genet Sel Evol 2020; 52:52. [PMID: 32887549 PMCID: PMC7487911 DOI: 10.1186/s12711-020-00571-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/21/2020] [Indexed: 02/01/2023] Open
Abstract
Background Thousands of years of natural and artificial selection have resulted in indigenous cattle breeds that are well-adapted to the environmental challenges of their local habitat and thereby are considered as valuable genetic resources. Understanding the genetic background of such adaptation processes can help us design effective breeding objectives to preserve local breeds and improve commercial cattle. To identify regions under putative selection, GGP HD 150 K single nucleotide polymorphism (SNP) arrays were used to genotype 106 individuals representing five Swedish breeds i.e. native to different regions and covering areas with a subarctic cold climate in the north and mountainous west, to those with a continental climate in the more densely populated south regions. Results Five statistics were incorporated within a framework, known as de-correlated composite of multiple signals (DCMS) to detect signatures of selection. The obtained p-values were adjusted for multiple testing (FDR < 5%), and significant genomic regions were identified. Annotation of genes in these regions revealed various verified and novel candidate genes that are associated with a diverse range of traits, including e.g. high altitude adaptation and response to hypoxia (DCAF8, PPP1R12A, SLC16A3, UCP2, UCP3, TIGAR), cold acclimation (AQP3, AQP7, HSPB8), body size and stature (PLAG1, KCNA6, NDUFA9, AKAP3, C5H12orf4, RAD51AP1, FGF6, TIGAR, CCND2, CSMD3), resistance to disease and bacterial infection (CHI3L2, GBP6, PPFIBP1, REP15, CYP4F2, TIGD2, PYURF, SLC10A2, FCHSD2, ARHGEF17, RELT, PRDM2, KDM5B), reproduction (PPP1R12A, ZFP36L2, CSPP1), milk yield and components (NPC1L1, NUDCD3, ACSS1, FCHSD2), growth and feed efficiency (TMEM68, TGS1, LYN, XKR4, FOXA2, GBP2, GBP5, FGD6), and polled phenotype (URB1, EVA1C). Conclusions We identified genomic regions that may provide background knowledge to understand the mechanisms that are involved in economic traits and adaptation to cold climate in cattle. Incorporating p-values of different statistics in a single DCMS framework may help select and prioritize candidate genes for further analyses.
Collapse
Affiliation(s)
- Seyed Mohammad Ghoreishifar
- Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj, 31587-11167, Iran
| | - Susanne Eriksson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, SE-75007, Uppsala, Sweden.
| | - Anna M Johansson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, SE-75007, Uppsala, Sweden
| | - Majid Khansefid
- AgriBio Centre for AgriBioscience, Agriculture Victoria, Bundoora, VIC, 3083, Australia
| | - Sima Moghaddaszadeh-Ahrabi
- Department of Animal Science, Faculty of Agriculture and Natural Resources, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Nahid Parna
- Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj, 31587-11167, Iran
| | - Pourya Davoudi
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, B2N5E3, Canada
| | - Arash Javanmard
- Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| |
Collapse
|
26
|
Uray K, Major E, Lontay B. MicroRNA Regulatory Pathways in the Control of the Actin-Myosin Cytoskeleton. Cells 2020; 9:E1649. [PMID: 32660059 PMCID: PMC7408560 DOI: 10.3390/cells9071649] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are key modulators of post-transcriptional gene regulation in a plethora of processes, including actin-myosin cytoskeleton dynamics. Recent evidence points to the widespread effects of miRNAs on actin-myosin cytoskeleton dynamics, either directly on the expression of actin and myosin genes or indirectly on the diverse signaling cascades modulating cytoskeletal arrangement. Furthermore, studies from various human models indicate that miRNAs contribute to the development of various human disorders. The potentially huge impact of miRNA-based mechanisms on cytoskeletal elements is just starting to be recognized. In this review, we summarize recent knowledge about the importance of microRNA modulation of the actin-myosin cytoskeleton affecting physiological processes, including cardiovascular function, hematopoiesis, podocyte physiology, and osteogenesis.
Collapse
Affiliation(s)
- Karen Uray
- Correspondence: (K.U.); (B.L.); Tel.: +36-52-412345 (K.U. & B.L.)
| | | | - Beata Lontay
- Correspondence: (K.U.); (B.L.); Tel.: +36-52-412345 (K.U. & B.L.)
| |
Collapse
|
27
|
Su M, Fan S, Ling Z, Fan X, Xia L, Liu Y, Li S, Zhang Y, Zeng Z, Tang WH. Restoring the Platelet miR-223 by Calpain Inhibition Alleviates the Neointimal Hyperplasia in Diabetes. Front Physiol 2020; 11:742. [PMID: 32733269 PMCID: PMC7359912 DOI: 10.3389/fphys.2020.00742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022] Open
Abstract
Platelet hyperactivity is the hallmark of diabetes, and platelet activation plays a crucial role in diabetic vascular complications. Recent studies have shown that upon activation, platelet-derived miRNAs are incorporated into vascular smooth muscle cells (VSMCs), regulating the phenotypic switch of VSMC. Under diabetes, miRNA deficiency in platelets fails to regulate the VSMC phenotypic switch. Therefore, manipulation of platelet-derived miRNAs expression may provide therapeutic option for diabetic vascular complications. We seek to investigate the effect of calpeptin (calpain inhibitor) on the expression of miRNAs in diabetic platelets, and elucidate the downstream signaling pathway involved in protecting from neointimal formation in diabetic mice with femoral wire injury model. Using human cell and platelet coculture, we demonstrate that diabetic platelet deficient of miR-223 fails to suppress VSMC proliferation, while overexpression of miR-223 in diabetic platelets suppressed the proliferation of VSMC to protect intimal hyperplasia. Mechanistically, miR-223 directly targets the insulin-like growth factor-1 receptor (IGF-1R), which inhibits the phosphorylation of GSK3β and activates the phosphorylation of AMPK, resulting in reduced VSMC dedifferentiation and proliferation. Using a murine model of vascular injury, we show that calpeptin restores the platelet expression of miR-223 in diabetes, and the horizontal transfer of platelet miR-223 into VSMCs inhibits VSMC proliferation in the injured artery by targeting the expression of IGF-1R. Our data present that the platelet-derived miR-223 suppressed VSMC proliferation via the regulation miR-223/IGF-1R/AMPK signaling pathways, and inhibition of calpain alleviates neointimal formation by restoring the expression of miR-223 in diabetic platelet.
Collapse
Affiliation(s)
- Meiling Su
- Joint Program in Cardiovascular Medicine, Affiliated Guangzhou Women and Children's Medical Centre, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Shunyang Fan
- Heart Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenwei Ling
- Joint Program in Cardiovascular Medicine, Affiliated Guangzhou Women and Children's Medical Centre, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Xuejiao Fan
- Joint Program in Cardiovascular Medicine, Affiliated Guangzhou Women and Children's Medical Centre, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Luoxing Xia
- Joint Program in Cardiovascular Medicine, Affiliated Guangzhou Women and Children's Medical Centre, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Yingying Liu
- Joint Program in Cardiovascular Medicine, Affiliated Guangzhou Women and Children's Medical Centre, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Shaoying Li
- Joint Program in Cardiovascular Medicine, Affiliated Guangzhou Women and Children's Medical Centre, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Yuan Zhang
- Joint Program in Cardiovascular Medicine, Affiliated Guangzhou Women and Children's Medical Centre, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Zhi Zeng
- Joint Program in Cardiovascular Medicine, Affiliated Guangzhou Women and Children's Medical Centre, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Wai Ho Tang
- Joint Program in Cardiovascular Medicine, Affiliated Guangzhou Women and Children's Medical Centre, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
28
|
Li S, Feng Y, Huang Y, Liu Y, Wang Y, Liang Y, Zeng H, Qu H, Wei L. MiR-223-3p regulates cell viability, migration, invasion, and apoptosis of non-small cell lung cancer cells by targeting RHOB. Open Life Sci 2020; 15:389-399. [PMID: 33817227 PMCID: PMC7874547 DOI: 10.1515/biol-2020-0040] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/25/2020] [Accepted: 03/31/2020] [Indexed: 12/15/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer with a high fatality rate in men and women worldwide. Recently, microRNAs (miRNAs) have been reported to be diagnostic biomarkers and therapeutic targets in NSCLC. MiR-223-3p was proved to act as a promoter in NSCLC progression. However, the regulatory mechanism of miR-223-3p in NSCLC remains little known. This study aimed to explore the regulatory mechanism between miR-223-3p and its target gene Ras homolog family member B (RHOB) in NSCLC. The mRNA level of miR-223-3p and RHOB was measured by quantitative reverse transcription PCR. Furthermore, cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Flow cytometry was conducted to analyze cell apoptosis. Transwell assays and wound healing assay were employed to examine migration and invasion. The target relationship between miR-223-3p and RHOB was predicted by starBase online database and verified by dual-luciferase assay. The protein level of RHOB was tested by western blot. Our data suggested that miR-223-3p was upregulated in NSCLC tissues and cell lines and high level of miR-223-3p contributed to a poor survival in NSCLC patients. Knockdown of miR-223-3p exerted inhibitory effects on NSCLC cell viability, migration, and invasion and promotion effect on cell apoptosis. Furthermore, RHOB was directly targeted by miR-223-3p and constrained NSCLC progression. Moreover, knockdown of RHOB rescued the effect of anti-miR-223-3p on NSCLC progression. In vivo experiments indicated that miR-223-3p deletion suppressed tumor growth. MiR-223-3p could regulate the NSCLC cellular processes through targeting RHOB.
Collapse
Affiliation(s)
- Shufang Li
- Department of Respiratory Medicine, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Linxia Rd, Chengguan District, 730030, Lanzhou, China
| | - Yuping Feng
- Department of Emergency, Gansu Provincial Third People’s Hospital, 730000, Lanzhou, China
| | - Yuxia Huang
- Department of Respiratory Medicine, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Linxia Rd, Chengguan District, 730030, Lanzhou, China
| | - Yu Liu
- Department of Respiratory Medicine, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Linxia Rd, Chengguan District, 730030, Lanzhou, China
| | - Yanxi Wang
- Department of Respiratory Medicine, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Linxia Rd, Chengguan District, 730030, Lanzhou, China
| | - Yan Liang
- Department of Respiratory Medicine, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Linxia Rd, Chengguan District, 730030, Lanzhou, China
| | - Hui Zeng
- Department of Respiratory Medicine, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Linxia Rd, Chengguan District, 730030, Lanzhou, China
| | - Hong Qu
- Department of Respiratory Medicine, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Linxia Rd, Chengguan District, 730030, Lanzhou, China
| | - Ling Wei
- Department of Respiratory Medicine, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Linxia Rd, Chengguan District, 730030, Lanzhou, China
| |
Collapse
|
29
|
Moorthy S, Byfield FJ, Janmey PA, Klein EA. Matrix stiffness regulates endosomal escape of uropathogenic E. coli. Cell Microbiol 2020; 22:e13196. [PMID: 32083802 DOI: 10.1111/cmi.13196] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 01/05/2023]
Abstract
Uropathogenic E. coli (UPEC) infection in vivo is characterized by invasion of bladder umbrella epithelial cells followed by endosomal escape and proliferation in the cytoplasm to form intracellular bacterial communities. By contrast, UPEC infection in tissue culture models results in bacteria being trapped within Lamp1-positive endosomes where proliferation is limited. Pharmacological disruption of the actin cytoskeleton has been shown to facilitate UPEC endosomal escape in vitro and extracellular matrix stiffness is a well-characterized physiological regulator of actin dynamics; therefore, we hypothesized that substrate stiffness may play a role in UPEC endosomal escape. Using functionalized polyacrylamide substrates, we found that at physiological stiffness, UPEC escaped the endosome and proliferated rapidly in the cytoplasm of bladder epithelial cells. Dissection of the cytoskeletal signaling pathway demonstrated that inhibition of the Rho GTPase RhoB or its effector PRK1 was sufficient to increase cytoplasmic bacterial growth and that RhoB protein level was significantly reduced at physiological stiffness. Our data suggest that tissue stiffness is a critical regulator of intracellular bacterial growth. Due to the ease of doing genetic and pharmacological manipulations in cell culture, this model system may provide a useful tool for performing mechanistic studies on the intracellular life cycle of uropathogens.
Collapse
Affiliation(s)
- Sudha Moorthy
- Department of Biology, Rutgers University-Camden, Camden, New Jersey, USA
| | - Fitzroy J Byfield
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul A Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eric A Klein
- Department of Biology, Rutgers University-Camden, Camden, New Jersey, USA.,Center for Computational and Integrative Biology, Rutgers University-Camden, Camden, New Jersey, USA
| |
Collapse
|
30
|
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
|
31
|
Fan J, Fan X, Guang H, Shan X, Tian Q, Zhang F, Chen R, Ye F, Quan H, Zhang H, Ding L, Gan Z, Xue F, Wang Y, Mao S, Hu L, Gong Y. Upregulation of miR-335-3p by NF-κB Transcriptional Regulation Contributes to the Induction of Pulmonary Arterial Hypertension via APJ during Hypoxia. Int J Biol Sci 2020; 16:515-528. [PMID: 32015687 PMCID: PMC6990898 DOI: 10.7150/ijbs.34517] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a cardiopulmonary disease that can lead to heart failure and eventually death. MicroRNAs (miRs) play essential roles during PAH progression; however, their exact mechanism of action remains unclear. Apelin is a small bioactive peptide with a key protective function in the pathogenesis of PAH mediated by binding to the APJ gene. The aim of the present study was to investigate the role of miR-335-3p in chronic normobaric hypoxia (CNH)-induced PAH in mice and the potential underlying regulatory mechanism. Adult male C57BL/6 mice were exposed to normoxia (~21% O2) or CNH (~10% O2, 23 h/d) for 5 weeks. MiR-335-3p was significantly increased in lung tissue of CNH-induced PAH mice. Blocking miR-335-3p attenuated CNH-induced PAH and alleviated pulmonary vascular remodeling. Bioinformatics analysis and luciferase reporter assay indicated that nuclear factor-kappa beta (NF-κB) acted as a transcriptional regulator upstream of miR-335-3p. Pyrrolidine dithiocarbamate treatment reversed the CNH-induced increase in miR-335-3p expression and diminished CNH-induced PAH. Moreover, p50-/- mice were resistant to CNH-induced PAH. Finally, APJ was identified as a direct targeting gene downstream of miR-335-3p, and pharmacological activation of APJ by its ligand apelin-13 reduced CNH-induced PAH and improved pulmonary vascular remodeling. Our results indicate that NF-κB-mediated transcriptional upregulation of miR-335-3p contributes to the inhibition of APJ and induction of PAH during hypoxia; hence, miR-335-3p could be a potential therapeutic target for hypoxic PAH.
Collapse
Affiliation(s)
- Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Hui Guang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiaoqiong Shan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Qiuyun Tian
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Fukun Zhang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Ran Chen
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Fangzhou Ye
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Hui Quan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Haizeng Zhang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Lu Ding
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhuohui Gan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Feng Xue
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yongyu Wang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Sunzhong Mao
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Lianggang Hu
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| |
Collapse
|
32
|
Zhao M, Chen N, Li X, Lin L, Chen X. MiR-19a modulates hypoxia-mediated cell proliferation and migration via repressing PTEN in human pulmonary arterial smooth muscle. Life Sci 2019; 239:116928. [PMID: 31682848 DOI: 10.1016/j.lfs.2019.116928] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/23/2019] [Accepted: 10/01/2019] [Indexed: 12/16/2022]
Abstract
AIM The dysfunction of human pulmonary arterial smooth muscle cells (HPASMCs) has been suggested to participate in the pathophysiology of pulmonary arterial hypertension (PAH). This study determined miR-19a expression in hypoxia-induced HPASMCs and explored the mechanistic actions of miR-19a in hypoxia-induced HPASMC proliferation and migration. METHODS QRT-PCR and western blot assays respectively determined the mRNA and protein expression of miR-19a, phosphatase and tensin homolog (PTEN) and hypoxia-inducible factor-1 alpha (HIF-1α). In vitro functional assays determined HPASMC proliferation and migration, respectively. Luciferase reporter assay determined interaction between miR-19a and PTEN. The knockdown effects of miR-19a on PAH were confirmed in in vivo mice model. RESULTS Hypoxia treatment time-dependently up-regulated miR-19a expression and enhanced cell proliferation in HPASMCs. MiR-19a overexpression increased cell proliferation and migration of HPASMCs, while repression of miR-19a reduced cell proliferative and migratory potentials of hypoxia-treated HPASMCs. Bioinformatics analysis and luciferase reporter assay showed that PTEN 3' untranslated region was targeted by miR-19a, and miR-19a repressed the mRNA and protein expression of PTEN in HPASMCs. Further rescue studies revealed that miR-19a regulated proliferative and migratory potentials of hypoxia-treated HPASMCs via suppressing PTEN expression. In addition, HIF-1α was identified as one of the mediators for the hypoxia-induced aberrant expression levels of miR-19a and PTEN. MiR-19a overexpression enhanced PI3K/AKT signaling, which was attenuated by enforced expression of PTEN in HPASMCs. More importantly, knockdown of miR-19 attenuated the chronic hypoxia-induced PAH in in vivo mice model. CONCLUSION This study presented a novel mechanistic action of miR-19a-mediated cell proliferation and migration of HPASMCs.
Collapse
Affiliation(s)
- Mei Zhao
- Department of Pharmacy, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya City, Hainan Province, China.
| | - Ni Chen
- Department of Pharmacy, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China.
| | - Xuelian Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin City, Heilongjiang Province, China.
| | - Ling Lin
- Department of Cardiovascular Medicine, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya City, Hainan Province, China.
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, China.
| |
Collapse
|
33
|
Abstract
Introduction: Glaucoma is a group of progressive optic neuropathies in which elevated intraocular pressure (IOP) as a consequence of an increased aqueous humor (AH) outflow resistance, is the main and only clinically modifiable risk factor for its development and progression. Relaxing Trabecular meshwork (TM) tissue, Rho-Kinase (ROCK) inhibitors directly decrease resistance in the conventional AH outflow, thus resulting in a significant IOP-lowering effect. Areas covered: The progress made in the field of ROCK inhibitors for glaucoma treatment will be discussed, referring to the recent patent literature published mainly in the last 3 years. Development and last studies conducted on the recently approved ripasudil and netarsudil will be described, along with newly reported combinations with other antiglaucoma agents. New molecular entities as ROCK inhibitors will be reported as well as new biological approaches to affect the Rho/ROCK pathway. Expert opinion: With three drugs currently available on the market belonging to this class, ROCK inhibitors have been definitely validated as therapeutic agents for glaucoma treatment. The literature of the last 3 years confirmed the success of the soft-drug and bis-functional approaches in the design of ROCK inhibitors. However, few completely new molecular scaffolds have been reported.
Collapse
Affiliation(s)
- Emanuela Berrino
- NEUROFARBA Dept., Sezione di Scienze Farmaceutiche, University of Florence , Sesto Fiorentino (Florence) , Italy
| | - Claudiu T Supuran
- NEUROFARBA Dept., Sezione di Scienze Farmaceutiche, University of Florence , Sesto Fiorentino (Florence) , Italy
| |
Collapse
|
34
|
Gutierrez E, Cahatol I, Bailey CAR, Lafargue A, Zhang N, Song Y, Tian H, Zhang Y, Chan R, Gu K, Zhang ACC, Tang J, Liu C, Connis N, Dennis P, Zhang C. Regulation of RhoB Gene Expression during Tumorigenesis and Aging Process and Its Potential Applications in These Processes. Cancers (Basel) 2019; 11:cancers11060818. [PMID: 31200451 PMCID: PMC6627600 DOI: 10.3390/cancers11060818] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/01/2019] [Accepted: 06/06/2019] [Indexed: 12/12/2022] Open
Abstract
RhoB, a member of the Ras homolog gene family and GTPase, regulates intracellular signaling pathways by interfacing with epidermal growth factor receptor (EGFR), Ras, and phosphatidylinositol 3-kinase (PI3K)/Akt to modulate responses in cellular structure and function. Notably, the EGFR, Ras, and PI3K/Akt pathways can lead to downregulation of RhoB, while simultaneously being associated with an increased propensity for tumorigenesis. Functionally, RhoB, part of the Rho GTPase family, regulates intracellular signaling pathways by interfacing with EGFR, RAS, and PI3K/Akt/mammalian target of rapamycin (mTOR), and MYC pathways to modulate responses in cellular structure and function. Notably, the EGFR, Ras, and PI3K/Akt pathways can lead to downregulation of RhoB, while simultaneously being associated with an increased propensity for tumorigenesis. RHOB expression has a complex regulatory backdrop consisting of multiple histone deacetyltransferase (HDACs 1 and 6) and microRNA (miR-19a, -21, and -223)-mediated mechanisms of modifying expression. The interwoven nature of RhoB’s regulatory impact and cellular roles in regulating intracellular vesicle trafficking, cell motion, and the cell cycle lays the foundation for analyzing the link between loss of RhoB and tumorigenesis within the context of age-related decline in RhoB. RhoB appears to play a tissue-specific role in tumorigenesis, as such, uncovering and appreciating the potential for restoration of RHOB expression as a mechanism for cancer prevention or therapeutics serves as a practical application. An in-depth assessment of RhoB will serve as a springboard for investigating and characterizing this key component of numerous intracellular messaging and regulatory pathways that may hold the connection between aging and tumorigenesis.
Collapse
Affiliation(s)
- Eutiquio Gutierrez
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E 2nd Street, Pomona, CA 91766, USA.
- Department of Internal Medicine, Harbor-UCLA Medical Center, 1000 W Carson Street, Torrance, CA 90509, USA.
| | - Ian Cahatol
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E 2nd Street, Pomona, CA 91766, USA
- Department of Graduate Medical Education, Community Memorial Health System, 147 N Brent Street, Ventura, CA 93003, USA
| | - Cedric A R Bailey
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E 2nd Street, Pomona, CA 91766, USA
- Department of Pathology and Immunology, Washington University School of Medicine, 509 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Audrey Lafargue
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21231, USA
| | - Naming Zhang
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Ying Song
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Hongwei Tian
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Yizhi Zhang
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Ryan Chan
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Kevin Gu
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Angel C C Zhang
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - James Tang
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Chunshui Liu
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Nick Connis
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Phillip Dennis
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Chunyu Zhang
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| |
Collapse
|
35
|
Kang K, Huang L, Li Q, Liao X, Dang Q, Yang Y, Luo J, Zeng Y, Li L, Gou D. An improved Tet-on system in microRNA overexpression and CRISPR/Cas9-mediated gene editing. J Anim Sci Biotechnol 2019; 10:43. [PMID: 31198556 PMCID: PMC6556963 DOI: 10.1186/s40104-019-0354-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/12/2019] [Indexed: 11/10/2022] Open
Abstract
Background Tetracycline (Tet)-regulated expression system has become a widely applied tool to control gene activity. This study aimed to improve the Tet-on system with superior regulatory characteristics. Results By comprehensively comparing factors of transactivators, Tet-responsive elements (TREs), orientations of induced expression cassette, and promoters controlling the transactivator, we developed an optimal Tet-on system with enhanced inducible efficiency and lower leakiness. With the system, we successfully performed effective inducible and reversible expression of microRNA, and presented a more precise and easily reproducible fine-tuning for confirming the target of a miRNA. Finally, the system was applied in CRISPR/Cas9-mediated knockout of nuclear factor of activated T cells-5 (NFAT5), a protective transcription factor in cellular osmoregulation. Conclusions This study established an improved Tet-on system for powerful and stringent gene regulation in functional genetic studies. Electronic supplementary material The online version of this article (10.1186/s40104-019-0354-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kang Kang
- 1Department of Biochemistry and Molecular Biology, Carson International Cancer Center, Shenzhen University Health Sciences Center, Shenzhen, Guangdong 518060 People's Republic of China
| | - Lian Huang
- 2Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100 People's Republic of China
| | - Qing Li
- 3Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Xueyuan Ave 1066, Shenzhen, Guangdong 518060 People's Republic of China
| | - Xiaoyun Liao
- 3Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Xueyuan Ave 1066, Shenzhen, Guangdong 518060 People's Republic of China
| | - Quanjin Dang
- 3Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Xueyuan Ave 1066, Shenzhen, Guangdong 518060 People's Republic of China
| | - Yi Yang
- 1Department of Biochemistry and Molecular Biology, Carson International Cancer Center, Shenzhen University Health Sciences Center, Shenzhen, Guangdong 518060 People's Republic of China
| | - Jun Luo
- 2Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100 People's Republic of China
| | - Yan Zeng
- 3Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Xueyuan Ave 1066, Shenzhen, Guangdong 518060 People's Republic of China
| | - Li Li
- 3Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Xueyuan Ave 1066, Shenzhen, Guangdong 518060 People's Republic of China
| | - Deming Gou
- 1Department of Biochemistry and Molecular Biology, Carson International Cancer Center, Shenzhen University Health Sciences Center, Shenzhen, Guangdong 518060 People's Republic of China.,3Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Xueyuan Ave 1066, Shenzhen, Guangdong 518060 People's Republic of China
| |
Collapse
|
36
|
Edea Z, Dadi H, Dessie T, Kim KS. Genomic signatures of high-altitude adaptation in Ethiopian sheep populations. Genes Genomics 2019; 41:973-981. [PMID: 31119684 DOI: 10.1007/s13258-019-00820-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 04/11/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Ethiopian sheep populations such as Arsi-Bale, Horro and Adilo (long fat-tailed, LFT) inhabit mid to high-altitude areas; and Menz sheep (MZ, short fat-tailed) are adapted to cool sub-alpine environments. In contrast, Blackhead Somali sheep (BHS, fat-rumped) thrive well in arid and semi-arid areas characterized by high temperature and low precipitation. The genomic investigation of Ethiopian sheep populations may help to identify genes and biological pathways enable to adapt to the different ecological conditions. OBJECTIVE To uncover genomic regions and genes showing evidence of positive selection for altitude adaptation in Ethiopian sheep populations. METHODS A total of 72 animals inhabiting high-versus low-altitude environments were genotyped on an Ovine Infinium HD array (~ 600 K). Pairwise genetic differentiation (Fst) was calculated in sliding windows of 20 SNPs and the upper 1% smoothed Fst values were considered to represent positive selection signatures. Genes within < 25 kb of the most differentiated SNPs were considered as selection candidates. RESULTS Signatures of selection were detected in genes known to be associated high with altitude adaptation in MZ-BHS pair comparison (PPP1R12A, RELN, PARP2, and DNAH9) and in LFT-BHS pair comparison (VAV3, MSRB3,EIF2AK4, MET, and TACR1). The candidate genes (MITF, FGF5, MTOR, TRHDE, and TUBB3) associated with altitude adaptation and shared between the MZ-BHS and LTF-BHS pair comparisons were also detected as under selection. Further functional analyses reveal that the candidate genes were involved in biological processes and pathways relevant to adaptation under extreme altitudes, including respiratory system development and smoothened signaling pathway. CONCLUSION The results of the present study could aid in-depth understanding and exploitation of the underlying genetic mechanisms for sheep and other livestock species adaptation to high-altitude environments.
Collapse
Affiliation(s)
- Zewdu Edea
- Department of Animal Science, Chungbuk National University, Cheongju, Korea
| | - Hailu Dadi
- Addis Ababa Science and Technology University, P. O. Box 2490, Addis Ababa, Ethiopia
| | - Tadelle Dessie
- International Livestock Research Institute, Addis Ababa, Ethiopia
| | - Kwan-Suk Kim
- Department of Animal Science, Chungbuk National University, Cheongju, Korea.
| |
Collapse
|
37
|
Metzinger-Le Meuth V, Metzinger L. miR-223 and other miRNA's evaluation in chronic kidney disease: Innovative biomarkers and therapeutic tools. Noncoding RNA Res 2019; 4:30-35. [PMID: 30891535 PMCID: PMC6404357 DOI: 10.1016/j.ncrna.2019.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/04/2018] [Accepted: 01/18/2019] [Indexed: 02/06/2023] Open
Abstract
microRNAs (miRNAs) represent a recent breakthrough regarding gene expression regulation. They are instrumental players known to regulate post-transcriptional expression. miRNAs are short single stranded RNAs that base-pair with target mRNAs in specific regions mainly within their 3' untranslated region. We know now that miRNAs are involved in kidney physiopathology. We outline in this review the recent discoveries made on the roles of miRNAs in cellular and animal models of kidney disease but also in patients suffering from chronic kidney disease, acute kidney injury and so forth. miRNAs are potential innovative biomarkers in nephrology, but before being used in daily clinical routine, their expression in large cohorts will have to be assessed, and an effort will have to be made to standardize measurement methods and to select the most suitable tissues and biofluids. In addition to a putative role as biomarkers, up- or down-regulating miRNAs is a novel therapeutic approach to cure kidney disorders. We discuss in this review recent methods that could be used to deliver miRNAs in a specific and suitable way in kidney and other organs damaged by kidney failure such as the cardiovascular system.
Collapse
Affiliation(s)
- Valérie Metzinger-Le Meuth
- INSERM U1148, Laboratory for Vascular Translational Science (LVTS), UFR SMBH, Université Paris 13-Sorbonne Paris Cité, 93017 Bobigny Cedex, France
| | - Laurent Metzinger
- HEMATIM EA4666, C.U.R.S, Université de Picardie Jules Verne, 80025 Amiens Cedex 1, France
| |
Collapse
|
38
|
Yan Y, Lu K, Ye T, Zhang Z. MicroRNA‑223 attenuates LPS‑induced inflammation in an acute lung injury model via the NLRP3 inflammasome and TLR4/NF‑κB signaling pathway via RHOB. Int J Mol Med 2019; 43:1467-1477. [PMID: 30747229 PMCID: PMC6365085 DOI: 10.3892/ijmm.2019.4075] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 01/17/2019] [Indexed: 11/05/2022] Open
Abstract
Acute lung injury (ALI) and the more severe acute respiratory distress syndrome are common and complex inflammatory lung diseases. MicroRNAs (miRs) have emerged as novel gene regulatory molecules, serving a crucial role in a variety of complex diseases, including ALI. In the present study, the anti‑inflammatory action of miR‑223 on inflammation in ALI was demonstrated and the possible mechanism was further examined. In lipopolysaccharide‑induced ALI, the expression of miR‑223 was reduced compared with that in the control normal group. An in vitro model was used to analyze the effect of miR‑223 downregulation on an ALI model, which increased inflammation, and induced the activation of the NACHT, LRR and PYD domains‑containing protein 3 (NLRP3) inflammasome and Toll‑like receptor 4 (TLR4)/nuclear factor (NF)‑κB signaling pathway via rho‑related GTP‑binding protein RhoB (RHOB). In addition, the overexpression of miR‑223 reduced inflammation and suppressed the NLRP3 inflammasome and TLR4/NF‑κB signaling pathway via RHOB in the in vitro model. Furthermore, TLR4 inhibitor or NLRP3 inhibitor reduced the pro‑inflammatory effect of miR‑223 downregulation in ALI. In conclusion, the results of the present study indicated that miR‑223 functioned as a biological indicator by regulating inflammation in ALI, and may represent a novel potential therapeutic target and prognostic marker of ALI.
Collapse
Affiliation(s)
- Yurong Yan
- Shandong University, Jinan, Shandong 250012, P.R. China
| | - Kexin Lu
- Department of Obstetrics, Binzhou Medical University Hospital, Binzhou, Shandong 256600, P.R. China
| | - Ting Ye
- Department of Anesthesiology, Binzhou Medical University Hospital, Binzhou, Shandong 256600, P.R. China
| | - Zongwang Zhang
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
39
|
Qian Z, Li Y, Yang H, Chen J, Li X, Gou D. PDGFBB promotes proliferation and migration via regulating miR-1181/STAT3 axis in human pulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2018; 315:L965-L976. [DOI: 10.1152/ajplung.00224.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Platelet-derived growth factor (PDGF) can induce hyperproliferation of pulmonary artery smooth muscle cells (PASMCs), which is a key causative factor to the occurrence and progression of pulmonary arterial hypertension (PAH). We previously identified that miR-1181 is significantly downregulated by PDGFBB in human PASMCs. In this work, we further explore the function of miR-1181 and underlying regulatory mechanisms in PDGF-induced PASMCs. First, the expression pattern of miR-1181 was characterized under PDGFBB treatment, and PDGF receptor/PKCβ signaling was found to repress miR-1181 expression. Then, gain- and loss-of-function experiments were respectively conducted and revealed the prominent role of miR-1181 in inhibiting PASMC proliferation and migration. Flow cytometry analysis suggested that miR-1181 regulated the PASMC proliferation through influencing the cell cycle transition from G0/G1 to S phase. Moreover, we exhibited that miR-1181 targeting STAT3 formed a regulatory axis to modulate PASMC proliferation. Finally, serum miR-1181 expression was also observed to be reduced in adult and newborn patients with PAH. Overall, this study provides novel findings that the miR-1181/STAT3 axis mediated PDGFBB-induced dysfunction in human PASMCs, implying a potential use of miR-1181 as a therapeutic and diagnostic candidate for the vascular remodeling diseases.
Collapse
Affiliation(s)
- Zhengjiang Qian
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yanjiao Li
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Haiyang Yang
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jidong Chen
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Xiang Li
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Deming Gou
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| |
Collapse
|
40
|
Luna RCP, de Oliveira Y, Lisboa JVC, Chaves TR, de Araújo TAM, de Sousa EE, Miranda Neto M, Pirola L, Braga VA, de Brito Alves JL. Insights on the epigenetic mechanisms underlying pulmonary arterial hypertension. ACTA ACUST UNITED AC 2018; 51:e7437. [PMID: 30365723 PMCID: PMC6207290 DOI: 10.1590/1414-431x20187437] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 09/04/2018] [Indexed: 12/21/2022]
Abstract
Pulmonary arterial hypertension (PAH), characterized by localized increased
arterial blood pressure in the lungs, is a slow developing long-term disease
that can be fatal. PAH is characterized by inflammation, vascular tone
imbalance, pathological pulmonary vascular remodeling, and right-sided heart
failure. Current treatments for PAH are palliative and development of new
therapies is necessary. Recent and relevant studies have demonstrated that
epigenetic processes may exert key influences on the pathogenesis of PAH and may
be promising therapeutic targets in the prevention and/or cure of this
condition. The aim of the present mini-review is to summarize the occurrence of
epigenetic-based mechanisms in the context of PAH physiopathology, focusing on
the roles of DNA methylation, histone post-translational modifications and
non-coding RNAs. We also discuss the potential of epigenetic-based therapies for
PAH.
Collapse
Affiliation(s)
- R C P Luna
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, PB, Brasil
| | - Y de Oliveira
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, PB, Brasil
| | - J V C Lisboa
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, PB, Brasil
| | - T R Chaves
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, PB, Brasil
| | - T A M de Araújo
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, PB, Brasil
| | - E E de Sousa
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, PB, Brasil
| | - M Miranda Neto
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, PB, Brasil
| | - L Pirola
- INSERM U1060, Lyon 1 University, Oullins, France
| | - V A Braga
- Departamento de Biotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, PB, Brasil
| | - J L de Brito Alves
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, PB, Brasil.,Departamento de Biotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, PB, Brasil
| |
Collapse
|
41
|
Tang Q, Li MY, Su YF, Fu J, Zou ZY, Wang Y, Li SN. Absence of miR-223-3p ameliorates hypoxia-induced injury through repressing cardiomyocyte apoptosis and oxidative stress by targeting KLF15. Eur J Pharmacol 2018; 841:67-74. [PMID: 30336138 DOI: 10.1016/j.ejphar.2018.10.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 01/23/2023]
Abstract
Apoptosis of cardiomyocytes and oxidant stress are considered essential processes in the progression of cardiovascular diseases. A hypoxic stress which causes apoptosis of cardiomyocytes is the main problem in ischemic heart disease. The aim of the present study was to explore the functional role and potential mechanisms of miR-223-3p in hypoxia-induced cardiomyocyte apoptosis and oxidative stress. Here, we observed a increment of miR-223-3p level accompanied by the decrease of Krüppel-like zinc-finger transcription factor 15 (KLF15) expression in response to hypoxia. Additionally, absence of miR-223-3p manifestly dampened hypoxia-induced cardiomyocyte injury in H9c2 cells, including improving cell viability, attenuating the LDH leakage and preventing cardiomyocyte apoptosis accompanied by an increase in the expression of Bcl-2 and a decrease in the expression of Bax and C-caspase 3 in the setting of hypoxia. Moreover, depletion of miR-223-3p evidently retarded oxidant stress by inhibiting reactive oxygen species generation and lipid peroxidation, as well as enhancing antioxidant enzyme activity in H9c2 cells following exposure to hypoxia. More importantly, KLF15 was a direct and functional target of miR-223-3p. Further data validated that miR-223-3p negatively regulated the expression of KLF15. Mechanistically, deletion of KLF15 partly abrogated the suppressive effects of miR-223-3p deletion on hypoxia-induced cardiomyocyte apoptosis and oxidative stress. Taken all data together, our findings established that our study defines a novel mechanism by which miR-223-3p protects against cardiomyocyte apoptosis and oxidative stress by targeting KLF15, suggesting that the miR-223-3p/KLF15 may be a potential therapeutic target for ischemic heart conditions.
Collapse
Affiliation(s)
- Qing Tang
- Department of Emergency, Xi'an Children's Hospital, Xi'an 710003, Shaanxi, PR China
| | - Ming-Yue Li
- Department of Emergency, Xi'an Children's Hospital, Xi'an 710003, Shaanxi, PR China
| | - Yu-Fei Su
- Department of Emergency, Xi'an Children's Hospital, Xi'an 710003, Shaanxi, PR China
| | - Jia Fu
- Department of Infection, Xi'an Children's Hospital, Xi'an 710003, Shaanxi, PR China
| | - Zong-Yi Zou
- Department of Emergency, Xi'an Children's Hospital, Xi'an 710003, Shaanxi, PR China
| | - Yi Wang
- Department of Critical Care Medicine, Xi'an Children's Hospital, Xi'an 710003, Shaanxi, PR China
| | - Shao-Ning Li
- Department of Emergency, Xi'an Children's Hospital, Xi'an 710003, Shaanxi, PR China.
| |
Collapse
|
42
|
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]
|
43
|
Notch3/VEGF-A axis is involved in TAT-mediated proliferation of pulmonary artery smooth muscle cells: Implications for HIV-associated PAH. Cell Death Discov 2018; 4:22. [PMID: 30109141 PMCID: PMC6078940 DOI: 10.1038/s41420-018-0087-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 07/10/2018] [Indexed: 01/09/2023] Open
Abstract
The incidence of pulmonary arterial hypertension (PAH) is a significant co-morbidity observed in HIV (+) individuals. Pulmonary artery smooth muscle cells (PASMCs)—key components of the arterial vessel wall that regulate vessel diameter, demonstrate increased proliferation and hypertrophy in the lungs of HIV infected individuals, underscoring the role of these cells in the pathogenesis of HIV-associated PAH. While several pathways have been implicated in enhanced proliferation of PASMCs, detailed molecular mechanism(s) underlying HIV-associated PASMC proliferation still remain elusive. In the current study, we sought to investigate the effects HIV protein transactivator of transcription (TAT)-mediated proliferation on PASMCs. In agreement with earlier findings, our results also demonstrated TAT-mediated proliferation of human PASMCs. We identified activation of a novel Notch3 signaling pathway in TAT-mediated proliferation of PASMCs. Further validation of the Notch 3 pathway was demonstrated using both pharmacological (γ-secretase inhibitor, DAPT), as well as genetic approaches (Notch3 siRNA). Vascular endothelial growth factor A (VEGF-A) was identified as a novel downstream molecule that was induced following Notch activation. Findings from in vitro studies were further validated in archived simian immunodeficiency virus (SIV)-infected monkey lung tissues. There was increased activation of Notch3 signaling as well as enhanced expression of VEGF-A in the lungs of SIV-infected macaques compared with the lungs of SIV(−) controls. Taken together, we demonstrated that HIV-TAT increased the proliferation of PASMCs via the Notch3/VEGF-A axis. Targeting the Notch3/VEGF-A axis could thus be considered a potential therapeutic approach for the treatment of HIV-associated PAH.
Collapse
|
44
|
Dual strands of the miR-223 duplex (miR-223-5p and miR-223-3p) inhibit cancer cell aggressiveness: targeted genes are involved in bladder cancer pathogenesis. J Hum Genet 2018. [PMID: 29540855 DOI: 10.1038/s10038-018-0437-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Analyses of microRNA (miRNA) expression signatures obtained by RNA sequencing revealed that some passenger miRNAs (miR-144-5p, miR-145-3p, miR-149-3p, miR-150-3p, and miR-199a-3p) acted as anti-tumor miRNAs in several types of cancer cells. The involvement of passenger strands in the pathogenesis of human cancer is a novel concept. Based on the miRNA signature of bladder cancer (BC) obtained by RNA sequencing, we focused on both strands of the miR-223-duplex (miR-223-5p and miR-223-3p) and investigated their functional significance in BC cells. Ectopic expression of these miRNAs showed that both miR-223-3p (the guide strand) and miR-223-5p (the passenger strand) inhibited cancer cell migration and invasion of BC cells. The role of miR-223-5p (the passenger strand) has not been well studied. Combining gene expression studies and in silico database analyses, we demonstrated the presence of 20 putative target genes that could be regulated by miR-223-5p in BC cells. Among these targets, high expression of five genes (ANLN, INHBA, OIP5, CCNB1, and CDCA2) was significantly associated with poor prognosis of BC patients based on The Cancer Genome Atlas (TCGA) database. Moreover, we showed that a gene (ANLN) encoding a multifunctional actin-binding protein was directly regulated by miR-223-5p in BC cells. Overexpression of ANLN was observed in BC clinical specimens and high expression of ANLN was significantly associated with poor prognosis of BC patients. We suggest that studies of regulatory cancer networks, including the passenger strands of miRNAs, may provide new insights into the pathogenic mechanisms of BC.
Collapse
|
45
|
Li Y, Li L, Qian Z, Lin B, Chen J, Luo Y, Qu J, Raj JU, Gou D. Phosphatidylinositol 3-Kinase-DNA Methyltransferase 1-miR-1281-Histone Deacetylase 4 Regulatory Axis Mediates Platelet-Derived Growth Factor-Induced Proliferation and Migration of Pulmonary Artery Smooth Muscle Cells. J Am Heart Assoc 2018; 7:e007572. [PMID: 29514810 PMCID: PMC5907547 DOI: 10.1161/jaha.117.007572] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/03/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Platelet-derived growth factor BB, a potent mitogen of pulmonary artery smooth muscle cells (PASMCs), has been implicated in pulmonary arterial remodeling, which is a key pathogenic feature of pulmonary arterial hypertension. Previous microRNA profiling in platelet-derived growth factor BB-treated PASMCs found a significantly downregulated microRNA, miR-1281, but it has not been associated with any cellular function, and we investigated the possibility. METHODS AND RESULTS Real-time quantitative reverse transcription-polymerase chain reaction assay proved that downregulation of miR-1281 was a conserved phenomenon in human and rat PASMCs. Overexpression and inhibition of miR-1281 in PASMCs promoted and suppressed, respectively, the cell proliferation and migration. Bioinformatic prediction and 3'-untranslated region reporter assay identified histone deacetylase 4 to be a direct target of miR-1281. Supporting this, proliferation and migration assay demonstrated the cellular function of histone deacetylase 4 is inversely correlated with that of miR-1281. Mechanistically, it is found that platelet-derived growth factor BB activates the phosphatidylinositol 3-kinase pathway, which then induces the expression of DNA methyltransferase 1, leading to enhanced methylation of a flanking CpG island and repressed miR-1281 expression. Finally, a reduced miR-1281 level was consistently identified in hypoxic PASMCs in vitro, in pulmonary arteries of rats with monocrotaline-induced pulmonary arterial hypertension, and in serum of patients with coronary heart disease-pulmonary arterial hypertension. These data suggest that there may be a diagnostic and therapeutic use for miR-1281. CONCLUSIONS Herein, we report a novel regulatory axis, phosphatidylinositol 3-kinase-DNA methyltransferase 1-miR-1281-histone deacetylase 4, integrating multiple epigenetic regulators that participate in platelet-derived growth factor BB-stimulated PASMC proliferation and migration and pulmonary vascular remodeling.
Collapse
MESH Headings
- Animals
- Becaplermin/pharmacology
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- DNA (Cytosine-5-)-Methyltransferase 1/metabolism
- Disease Models, Animal
- HEK293 Cells
- Histone Deacetylases/genetics
- Histone Deacetylases/metabolism
- Humans
- Hypertension, Pulmonary/enzymology
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Male
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Monocrotaline
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Phosphatidylinositol 3-Kinase/metabolism
- Pulmonary Artery/enzymology
- Pulmonary Artery/pathology
- Rats, Sprague-Dawley
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction/drug effects
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Yanjiao Li
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | - Li Li
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Zhengjiang Qian
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Boya Lin
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Jidong Chen
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | - Yixuan Luo
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | - J Usha Raj
- Department of Pediatrics, University of Illinois at Chicago, IL
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
46
|
Chen J, Guo J, Cui X, Dai Y, Tang Z, Qu J, Raj JU, Hu Q, Gou D. The Long Noncoding RNA LnRPT Is Regulated by PDGF-BB and Modulates the Proliferation of Pulmonary Artery Smooth Muscle Cells. Am J Respir Cell Mol Biol 2018; 58:181-193. [DOI: 10.1165/rcmb.2017-0111oc] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Jidong Chen
- Shenzhen Key Laboratory of Microbial Genetic Engineering
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences, and
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - Jiao Guo
- Shenzhen Key Laboratory of Microbial Genetic Engineering
| | - Xiaolei Cui
- Shenzhen Key Laboratory of Microbial Genetic Engineering
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences, and
| | - Yan Dai
- Key Laboratory of Systems Biology, Chinese Academy of Science, Shanghai Institute for Biological Sciences, Shanghai, China
| | - Zhixiong Tang
- Shenzhen Key Laboratory of Microbial Genetic Engineering
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences, and
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - J. Usha Raj
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; and
| | - Qinghua Hu
- Department of Pathophysiology and
- Key Laboratory of Pulmonary Diseases of Ministry of Health, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences, and
| |
Collapse
|
47
|
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
|
48
|
Yu H, Xu M, Dong Y, Liu J, Li Y, Mao W, Wang J, Wang L. 1,25(OH) 2 D 3 attenuates pulmonary arterial hypertension via microRNA-204 mediated Tgfbr2/Smad signaling. Exp Cell Res 2018; 362:311-323. [DOI: 10.1016/j.yexcr.2017.11.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/16/2017] [Accepted: 11/25/2017] [Indexed: 12/27/2022]
|
49
|
Florijn BW, Bijkerk R, van der Veer EP, van Zonneveld AJ. Gender and cardiovascular disease: are sex-biased microRNA networks a driving force behind heart failure with preserved ejection fraction in women? Cardiovasc Res 2017; 114:210-225. [DOI: 10.1093/cvr/cvx223] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 11/23/2017] [Indexed: 01/08/2023] Open
Abstract
AbstractCardiovascular disease (CVD) is the primary cause of death among men and women worldwide. Nevertheless, our comprehension of how CVD progresses in women and elicits clinical outcomes is lacking, leading CVD to be under-diagnosed and under-treated in women. A clear example of this differential presentation of CVD pathophysiologies in females is the strikingly higher prevalence of heart failure with preserved ejection fraction (HFpEF). Women with a history of pre-eclampsia or those who present with co-morbidities such as obesity, hypertension, and diabetes mellitus are at increased risk of developing HFpEF. Long understood to be a critical CVD risk factor, our understanding of how gender differentially affects the development of CVD has been greatly expanded by extensive genomic and transcriptomic studies. These studies uncovered a pivotal role for differential microRNA (miRNA) expression in response to systemic inflammation, where their co-ordinated expression forms a post-transcriptional regulatory network that instigates microcirculation defects. Importantly, the potential sex-biased expression of the given miRNAs may explain sex-specific cardiovascular pathophysiologies in women, such as HFpEF. Sex-biased miRNAs are regulated by oestrogen (E2) in their transcription and processing or are expressed from loci on the X-chromosome due to incomplete X-chromosome inactivation. Interestingly, while E2-induced miRNAs predominantly appear to serve protective functions, it could be argued that many X-linked miRNAs have been found to challenge microvascular and myocardial integrity. Therefore, menopausal E2 deficiency, resulting in protective miRNA loss, and the augmentation of X-linked miRNA expression, may well contribute to the molecular mechanisms that underlie the female-specific cardiovascular aetiology in HFpEF.
Collapse
Affiliation(s)
- Barend W Florijn
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Roel Bijkerk
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Eric P van der Veer
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| |
Collapse
|
50
|
Ly6G+ neutrophil-derived miR-223 inhibits the NLRP3 inflammasome in mitochondrial DAMP-induced acute lung injury. Cell Death Dis 2017; 8:e3170. [PMID: 29144508 PMCID: PMC5775410 DOI: 10.1038/cddis.2017.549] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 12/19/2022]
Abstract
MicroRNA (miRNA) mediates RNA interference to regulate a variety of innate immune processes, but how miRNAs coordinate the mechanisms underlying acute lung injury/acute respiratory distress syndrome (ALI/ARDS) in patients with pulmonary inflammatory injury is still unknown. In this study, we demonstrated that miR-223 limits the number of Ly6G+ neutrophils and inhibits the activity of the NLRP3 inflammasome to alleviate ALI induced by mitochondrial damage-associated molecular patterns (DAMPs) (MTDs). miR-223 expression is increased in the lungs of MTD-induced mice or ARDS patients following trauma/transfusion or following the physiological remission of ALI/ARDS. miR-223−/+ mice exhibited more severe ALI and cytokine dysregulation. Other studies have shown that MTD-induced increases in miR-223 expression are mainly contributed by Ly6G+ neutrophils from the haematopoietic system. miR-223 blocks bone marrow-derived Ly6G+ neutrophil differentiation and inhibits peripheral cytokine release. In addition, MTD-induced miR-223 expression activates a negative feedback pathway that targets the inhibition of NLRP3 expression and IL-1β release; therefore, miR-223 deficiency can lead to the sustained activation of NLRP3-IL-1β. Finally, elimination of peripheral Ly6G+ neutrophils and pharmacological blockade of the miR-223–NLRP3–IL-1β signalling axis could alleviate MTD-induced ALI. In summary, miR-223 is essential for regulating the pathogenesis of DAMP-induced ALI.
Collapse
|