51
|
The role of microRNAs in diseases and related signaling pathways. Mol Biol Rep 2021; 49:6789-6801. [PMID: 34718938 DOI: 10.1007/s11033-021-06725-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/27/2021] [Indexed: 10/19/2022]
Abstract
MicroRNAs (miRNAs) are epigenetic regulators of the gene expression and act through posttranslational modification. They bind to 3'-UTR of target mRNAs to inhibit translation or increase the degradation mRNA in many tissues. Any alteration in the level of miRNA expression in many human diseases indicates their involvement in the pathogenesis of many diseases. On the other hand, the regulation of the signaling pathways is necessary for the maintenance of natural and physiological characteristics of any cell. It is worth mentioning that dysfunction of the signaling pathways manifests itself as a disorder or disease. The significant evidence report that miRNAs regulate the several signaling pathways in many diseases. Base on previous studies, miRNAs can be used for therapeutic or diagnostic purposes. According to the important role of miRNAs on the cell signaling pathways, this article reviews miRNAs involvement in incidence of diseases by changing signaling pathways.
Collapse
|
52
|
Fan P, Zhang L, Cheng T, Wang J, Zhou J, Zhao L, Hua C, Xia Q. MiR-590-5p inhibits pathological hypertrophy mediated heart failure by targeting RTN4. J Mol Histol 2021; 52:955-964. [PMID: 34406553 DOI: 10.1007/s10735-021-10009-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/08/2021] [Indexed: 10/20/2022]
Abstract
Heart failure (HF) is a rising epidemic and public health burden in modern society. It is of great need to find new biomarkers to ensure a timely diagnosis and to improve treatment and prognosis of the disease. The mouse model of HF was established by thoracic aortic constriction. Color Doppler ultrasound was performed to detect left ventricular end-diastolic diameter. Hematoxylin and eosin staining was conducted to observe the pathological changes of mouse myocardium. The RT-qPCR analysis was performed to detect miR-590-5p and RTN4 expression levels. Western blot was conducted to detect protein levels of the indicated genes. We found that the expression of miR-590-5p was downregulated in cardiac tissues of HF mice. Injection of AAV-miR-590-5p attenuated myocardium hypertrophy and myocyte apoptosis. Additionally, miR-590-5p overexpression promoted viability, inhibited apoptosis, and decreased ANF, BNP and beta-MHC protein levels in H9c2 cell. Mechanistically, miR-590-5p binds to RTN4 3'-untranslated region, as predicted by starBase online database and evidenced by luciferase reporter assay. Furthermore, miR-590-5p negatively regulates RTN4 mRNA expression and suppresses its translation. The final rescue experiments revealed that miR-590-5p modulated cardiomyocyte phenotypes by binding to RTN4. In conclusion, miR-590-5p modulates myocardium hypertrophy and myocyte apoptosis in HF by downregulating RTN4.
Collapse
Affiliation(s)
- Ping Fan
- Department of Cardiovascular Medicine, The Air Force Hospital From Eastern Theater, Qinhuai District, No. 1, Malu Street, Nanjing, 210001, Jiangsu, China
| | - Likun Zhang
- Department of Cardiovascular Medicine, The Air Force Hospital From Eastern Theater, Qinhuai District, No. 1, Malu Street, Nanjing, 210001, Jiangsu, China
| | - Tianyu Cheng
- Department of Cardiovascular Medicine, The Air Force Hospital From Eastern Theater, Qinhuai District, No. 1, Malu Street, Nanjing, 210001, Jiangsu, China
| | - Jing Wang
- Department of Cardiovascular Medicine, The Air Force Hospital From Eastern Theater, Qinhuai District, No. 1, Malu Street, Nanjing, 210001, Jiangsu, China
- Department of General Practice, Confucius Temple Community Health Service Center, Nanjing, 210001, Jiangsu, China
| | - Junyun Zhou
- Department of Cardiovascular Medicine, The Air Force Hospital From Eastern Theater, Qinhuai District, No. 1, Malu Street, Nanjing, 210001, Jiangsu, China
| | - Li Zhao
- Department of Cardiovascular Medicine, The Air Force Hospital From Eastern Theater, Qinhuai District, No. 1, Malu Street, Nanjing, 210001, Jiangsu, China
| | - Cuie Hua
- Department of Cardiovascular Medicine, The Air Force Hospital From Eastern Theater, Qinhuai District, No. 1, Malu Street, Nanjing, 210001, Jiangsu, China.
| | - Quan Xia
- Department of Cardiovascular Medicine, The Air Force Hospital From Eastern Theater, Qinhuai District, No. 1, Malu Street, Nanjing, 210001, Jiangsu, China.
| |
Collapse
|
53
|
Klionsky DJ, Petroni G, Amaravadi RK, Baehrecke EH, Ballabio A, Boya P, Bravo‐San Pedro JM, Cadwell K, Cecconi F, Choi AMK, Choi ME, Chu CT, Codogno P, Colombo M, Cuervo AM, Deretic V, Dikic I, Elazar Z, Eskelinen E, Fimia GM, Gewirtz DA, Green DR, Hansen M, Jäättelä M, Johansen T, Juhász G, Karantza V, Kraft C, Kroemer G, Ktistakis NT, Kumar S, Lopez‐Otin C, Macleod KF, Madeo F, Martinez J, Meléndez A, Mizushima N, Münz C, Penninger JM, Perera R, Piacentini M, Reggiori F, Rubinsztein DC, Ryan K, Sadoshima J, Santambrogio L, Scorrano L, Simon H, Simon AK, Simonsen A, Stolz A, Tavernarakis N, Tooze SA, Yoshimori T, Yuan J, Yue Z, Zhong Q, Galluzzi L, Pietrocola F. Autophagy in major human diseases. EMBO J 2021; 40:e108863. [PMID: 34459017 PMCID: PMC8488577 DOI: 10.15252/embj.2021108863] [Citation(s) in RCA: 901] [Impact Index Per Article: 225.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a core molecular pathway for the preservation of cellular and organismal homeostasis. Pharmacological and genetic interventions impairing autophagy responses promote or aggravate disease in a plethora of experimental models. Consistently, mutations in autophagy-related processes cause severe human pathologies. Here, we review and discuss preclinical data linking autophagy dysfunction to the pathogenesis of major human disorders including cancer as well as cardiovascular, neurodegenerative, metabolic, pulmonary, renal, infectious, musculoskeletal, and ocular disorders.
Collapse
Affiliation(s)
| | - Giulia Petroni
- Department of Radiation OncologyWeill Cornell Medical CollegeNew YorkNYUSA
| | - Ravi K Amaravadi
- Department of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- Abramson Cancer CenterUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Andrea Ballabio
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesSection of PediatricsFederico II UniversityNaplesItaly
- Department of Molecular and Human GeneticsBaylor College of Medicine, and Jan and Dan Duncan Neurological Research InstituteTexas Children HospitalHoustonTXUSA
| | - Patricia Boya
- Margarita Salas Center for Biological ResearchSpanish National Research CouncilMadridSpain
| | - José Manuel Bravo‐San Pedro
- Faculty of MedicineDepartment Section of PhysiologyComplutense University of MadridMadridSpain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball InstituteNew York University Grossman School of MedicineNew YorkNYUSA
- Department of MicrobiologyNew York University Grossman School of MedicineNew YorkNYUSA
- Division of Gastroenterology and HepatologyDepartment of MedicineNew York University Langone HealthNew YorkNYUSA
| | - Francesco Cecconi
- Cell Stress and Survival UnitCenter for Autophagy, Recycling and Disease (CARD)Danish Cancer Society Research CenterCopenhagenDenmark
- Department of Pediatric Onco‐Hematology and Cell and Gene TherapyIRCCS Bambino Gesù Children's HospitalRomeItaly
- Department of BiologyUniversity of Rome ‘Tor Vergata’RomeItaly
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care MedicineJoan and Sanford I. Weill Department of MedicineWeill Cornell MedicineNew YorkNYUSA
- New York‐Presbyterian HospitalWeill Cornell MedicineNew YorkNYUSA
| | - Mary E Choi
- New York‐Presbyterian HospitalWeill Cornell MedicineNew YorkNYUSA
- Division of Nephrology and HypertensionJoan and Sanford I. Weill Department of MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Charleen T Chu
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Patrice Codogno
- Institut Necker‐Enfants MaladesINSERM U1151‐CNRS UMR 8253ParisFrance
- Université de ParisParisFrance
| | - Maria Isabel Colombo
- Laboratorio de Mecanismos Moleculares Implicados en el Tráfico Vesicular y la Autofagia‐Instituto de Histología y Embriología (IHEM)‐Universidad Nacional de CuyoCONICET‐ Facultad de Ciencias MédicasMendozaArgentina
| | - Ana Maria Cuervo
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineBronxNYUSA
- Institute for Aging StudiesAlbert Einstein College of MedicineBronxNYUSA
| | - Vojo Deretic
- Autophagy Inflammation and Metabolism (AIMCenter of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Ivan Dikic
- Institute of Biochemistry IISchool of MedicineGoethe UniversityFrankfurt, Frankfurt am MainGermany
- Buchmann Institute for Molecular Life SciencesGoethe UniversityFrankfurt, Frankfurt am MainGermany
| | - Zvulun Elazar
- Department of Biomolecular SciencesThe Weizmann Institute of ScienceRehovotIsrael
| | | | - Gian Maria Fimia
- Department of Molecular MedicineSapienza University of RomeRomeItaly
- Department of EpidemiologyPreclinical Research, and Advanced DiagnosticsNational Institute for Infectious Diseases ‘L. Spallanzani’ IRCCSRomeItaly
| | - David A Gewirtz
- Department of Pharmacology and ToxicologySchool of MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Douglas R Green
- Department of ImmunologySt. Jude Children's Research HospitalMemphisTNUSA
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery InstituteProgram of DevelopmentAging, and RegenerationLa JollaCAUSA
| | - Marja Jäättelä
- Cell Death and MetabolismCenter for Autophagy, Recycling & DiseaseDanish Cancer Society Research CenterCopenhagenDenmark
- Department of Cellular and Molecular MedicineFaculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Terje Johansen
- Department of Medical BiologyMolecular Cancer Research GroupUniversity of Tromsø—The Arctic University of NorwayTromsøNorway
| | - Gábor Juhász
- Institute of GeneticsBiological Research CenterSzegedHungary
- Department of Anatomy, Cell and Developmental BiologyEötvös Loránd UniversityBudapestHungary
| | | | - Claudine Kraft
- Institute of Biochemistry and Molecular BiologyZBMZFaculty of MedicineUniversity of FreiburgFreiburgGermany
- CIBSS ‐ Centre for Integrative Biological Signalling StudiesUniversity of FreiburgFreiburgGermany
| | - Guido Kroemer
- Centre de Recherche des CordeliersEquipe Labellisée par la Ligue Contre le CancerUniversité de ParisSorbonne UniversitéInserm U1138Institut Universitaire de FranceParisFrance
- Metabolomics and Cell Biology PlatformsInstitut Gustave RoussyVillejuifFrance
- Pôle de BiologieHôpital Européen Georges PompidouAP‐HPParisFrance
- Suzhou Institute for Systems MedicineChinese Academy of Medical SciencesSuzhouChina
- Karolinska InstituteDepartment of Women's and Children's HealthKarolinska University HospitalStockholmSweden
| | | | - Sharad Kumar
- Centre for Cancer BiologyUniversity of South AustraliaAdelaideSAAustralia
- Faculty of Health and Medical SciencesUniversity of AdelaideAdelaideSAAustralia
| | - Carlos Lopez‐Otin
- Departamento de Bioquímica y Biología MolecularFacultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA)Universidad de OviedoOviedoSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Kay F Macleod
- The Ben May Department for Cancer ResearchThe Gordon Center for Integrative SciencesW‐338The University of ChicagoChicagoILUSA
- The University of ChicagoChicagoILUSA
| | - Frank Madeo
- Institute of Molecular BiosciencesNAWI GrazUniversity of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
- Field of Excellence BioHealth – University of GrazGrazAustria
| | - Jennifer Martinez
- Immunity, Inflammation and Disease LaboratoryNational Institute of Environmental Health SciencesNIHResearch Triangle ParkNCUSA
| | - Alicia Meléndez
- Biology Department, Queens CollegeCity University of New YorkFlushingNYUSA
- The Graduate Center Biology and Biochemistry PhD Programs of the City University of New YorkNew YorkNYUSA
| | - Noboru Mizushima
- Department of Biochemistry and Molecular BiologyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Christian Münz
- Viral ImmunobiologyInstitute of Experimental ImmunologyUniversity of ZurichZurichSwitzerland
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA)Vienna BioCenter (VBC)ViennaAustria
- Department of Medical GeneticsLife Sciences InstituteUniversity of British ColumbiaVancouverBCCanada
| | - Rushika M Perera
- Department of AnatomyUniversity of California, San FranciscoSan FranciscoCAUSA
- Department of PathologyUniversity of California, San FranciscoSan FranciscoCAUSA
- Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Mauro Piacentini
- Department of BiologyUniversity of Rome “Tor Vergata”RomeItaly
- Laboratory of Molecular MedicineInstitute of Cytology Russian Academy of ScienceSaint PetersburgRussia
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells & SystemsMolecular Cell Biology SectionUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - David C Rubinsztein
- Department of Medical GeneticsCambridge Institute for Medical ResearchUniversity of CambridgeCambridgeUK
- UK Dementia Research InstituteUniversity of CambridgeCambridgeUK
| | - Kevin M Ryan
- Cancer Research UK Beatson InstituteGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular MedicineCardiovascular Research InstituteRutgers New Jersey Medical SchoolNewarkNJUSA
| | - Laura Santambrogio
- Department of Radiation OncologyWeill Cornell Medical CollegeNew YorkNYUSA
- Sandra and Edward Meyer Cancer CenterNew YorkNYUSA
- Caryl and Israel Englander Institute for Precision MedicineNew YorkNYUSA
| | - Luca Scorrano
- Istituto Veneto di Medicina MolecolarePadovaItaly
- Department of BiologyUniversity of PadovaPadovaItaly
| | - Hans‐Uwe Simon
- Institute of PharmacologyUniversity of BernBernSwitzerland
- Department of Clinical Immunology and AllergologySechenov UniversityMoscowRussia
- Laboratory of Molecular ImmunologyInstitute of Fundamental Medicine and BiologyKazan Federal UniversityKazanRussia
| | | | - Anne Simonsen
- Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
- Centre for Cancer Cell ReprogrammingInstitute of Clinical MedicineUniversity of OsloOsloNorway
- Department of Molecular Cell BiologyInstitute for Cancer ResearchOslo University Hospital MontebelloOsloNorway
| | - Alexandra Stolz
- Institute of Biochemistry IISchool of MedicineGoethe UniversityFrankfurt, Frankfurt am MainGermany
- Buchmann Institute for Molecular Life SciencesGoethe UniversityFrankfurt, Frankfurt am MainGermany
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and BiotechnologyFoundation for Research and Technology‐HellasHeraklion, CreteGreece
- Department of Basic SciencesSchool of MedicineUniversity of CreteHeraklion, CreteGreece
| | - Sharon A Tooze
- Molecular Cell Biology of AutophagyThe Francis Crick InstituteLondonUK
| | - Tamotsu Yoshimori
- Department of GeneticsGraduate School of MedicineOsaka UniversitySuitaJapan
- Department of Intracellular Membrane DynamicsGraduate School of Frontier BiosciencesOsaka UniversitySuitaJapan
- Integrated Frontier Research for Medical Science DivisionInstitute for Open and Transdisciplinary Research Initiatives (OTRI)Osaka UniversitySuitaJapan
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryChinese Academy of SciencesShanghaiChina
- Department of Cell BiologyHarvard Medical SchoolBostonMAUSA
| | - Zhenyu Yue
- Department of NeurologyFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationDepartment of PathophysiologyShanghai Jiao Tong University School of Medicine (SJTU‐SM)ShanghaiChina
| | - Lorenzo Galluzzi
- Department of Radiation OncologyWeill Cornell Medical CollegeNew YorkNYUSA
- Sandra and Edward Meyer Cancer CenterNew YorkNYUSA
- Caryl and Israel Englander Institute for Precision MedicineNew YorkNYUSA
- Department of DermatologyYale School of MedicineNew HavenCTUSA
- Université de ParisParisFrance
| | | |
Collapse
|
54
|
Marracino L, Fortini F, Bouhamida E, Camponogara F, Severi P, Mazzoni E, Patergnani S, D’Aniello E, Campana R, Pinton P, Martini F, Tognon M, Campo G, Ferrari R, Vieceli Dalla Sega F, Rizzo P. Adding a "Notch" to Cardiovascular Disease Therapeutics: A MicroRNA-Based Approach. Front Cell Dev Biol 2021; 9:695114. [PMID: 34527667 PMCID: PMC8435685 DOI: 10.3389/fcell.2021.695114] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
Dysregulation of the Notch pathway is implicated in the pathophysiology of cardiovascular diseases (CVDs), but, as of today, therapies based on the re-establishing the physiological levels of Notch in the heart and vessels are not available. A possible reason is the context-dependent role of Notch in the cardiovascular system, which would require a finely tuned, cell-specific approach. MicroRNAs (miRNAs) are short functional endogenous, non-coding RNA sequences able to regulate gene expression at post-transcriptional levels influencing most, if not all, biological processes. Dysregulation of miRNAs expression is implicated in the molecular mechanisms underlying many CVDs. Notch is regulated and regulates a large number of miRNAs expressed in the cardiovascular system and, thus, targeting these miRNAs could represent an avenue to be explored to target Notch for CVDs. In this Review, we provide an overview of both established and potential, based on evidence in other pathologies, crosstalks between miRNAs and Notch in cellular processes underlying atherosclerosis, myocardial ischemia, heart failure, calcification of aortic valve, and arrhythmias. We also discuss the potential advantages, as well as the challenges, of using miRNAs for a Notch-based approach for the diagnosis and treatment of the most common CVDs.
Collapse
Affiliation(s)
- Luisa Marracino
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | | | - Esmaa Bouhamida
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesca Camponogara
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Paolo Severi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Elisa Mazzoni
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Simone Patergnani
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Emanuele D’Aniello
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Roberta Campana
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
| | | | - Paola Rizzo
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
| |
Collapse
|
55
|
Feng H, Xie B, Zhang Z, Yan J, Cheng M, Zhou Y. MiR-135a Protects against Myocardial Injury by Targeting TLR4. Chem Pharm Bull (Tokyo) 2021; 69:529-536. [PMID: 34078799 DOI: 10.1248/cpb.c20-01003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Emerging evidence highlights the importance of microRNAs (miRNAs) as functional regulators in cardiovascular disease. This study aimed to investigate the functional significance of miR-135a in the regulation of cardiac injury after isoprenaline (ISO) stimulation and the underlying mechanisms of its effects. Murine models with cardiac-specific overexpression of miR-135a were constructed with an adeno-associated virus expression system. The cardiac injury model was induced by ISO injection (60 mg/kg per day for 14 d). In vitro, we used H9c2 cells to establish a cell injury model by ISO stimulation (10 µM). The results indicated that miR-135a was increased during days 0-6 of ISO injection and was then downregulated during days 8-14 of ISO injection. The expression of miR-135a was consistent with the in vivo findings. Moreover, mice with cardiac overexpression of miR-135a exhibited reduced cardiac fibrosis, lactate dehydrogenase levels, Troponin I, inflammatory response and apoptosis. Overexpression of miR-135a also ameliorated cardiac dysfunction induced by ISO. MiR-135 overexpression in H9c2 cells increased cell viability and decreased cell apoptosis and inflammation in response to ISO. Conversely, miR-135 silencing in H9c2 cells decreased cell viability and increased cell apoptosis and inflammation in response to ISO. Mechanistically, we found that miR-135a negatively regulated toll-like receptor 4 (TLR4), which was confirmed by luciferase assay. Furthermore, the TLR4 inhibitor eritoran abolished the adverse effect of miR-135 silencing. Overall, miR-135a promotes ISO-induced cardiac injury by inhibiting the TLR4 pathway. MiR-135a may be a therapeutic agent for cardiac injury.
Collapse
Affiliation(s)
- Hui Feng
- Department of Cardiology, The First Affiliated Hospital of Soochow University.,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University
| | - Bing Xie
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University
| | - Zhuoqi Zhang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University
| | - Jun Yan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University
| | - Mingyue Cheng
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University
| | - Yafeng Zhou
- Department of Cardiology, The First Affiliated Hospital of Soochow University
| |
Collapse
|
56
|
Fan Y, Shao J, Wei S, Song C, Li Y, Jiang S. Self-eating and Heart: The Emerging Roles of Autophagy in Calcific Aortic Valve Disease. Aging Dis 2021; 12:1287-1303. [PMID: 34341709 PMCID: PMC8279526 DOI: 10.14336/ad.2021.0101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 01/01/2021] [Indexed: 12/16/2022] Open
Abstract
Autophagy is a self-degradative pathway by which subcellular elements are broken down intracellularly to maintain cellular homeostasis. Cardiac autophagy commonly decreases with aging and is accompanied by the accumulation of misfolded proteins and dysfunctional organelles, which are undesirable to the cell. Reduction of autophagy over time leads to aging-related cardiac dysfunction and is inversely related to longevity. However, despite the increasing interest in autophagy in cardiac diseases and aging, the process remains an undervalued and disregarded object in calcific valvular disease. Neither the nature through which autophagy is triggered nor the interplay between autophagic machinery and targeted molecules during aortic valve calcification are fully understood. Recently, the upregulation of autophagy has been shown to result in cardioprotective effects against cell death as well as its origin. Here, we review the evidence that shows how autophagy can be both beneficial and detrimental as it pertains to aortic valve calcification in the heart.
Collapse
Affiliation(s)
- Yunlong Fan
- 1Medical School of Chinese PLA, Beijing 100853, China.,2Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Jiakang Shao
- 1Medical School of Chinese PLA, Beijing 100853, China
| | - Shixiong Wei
- 1Medical School of Chinese PLA, Beijing 100853, China.,2Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Chao Song
- 1Medical School of Chinese PLA, Beijing 100853, China.,2Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Yanan Li
- 1Medical School of Chinese PLA, Beijing 100853, China
| | - Shengli Jiang
- 1Medical School of Chinese PLA, Beijing 100853, China.,2Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
57
|
Du H, Zhao Y, Li H, Wang DW, Chen C. Roles of MicroRNAs in Glucose and Lipid Metabolism in the Heart. Front Cardiovasc Med 2021; 8:716213. [PMID: 34368265 PMCID: PMC8339264 DOI: 10.3389/fcvm.2021.716213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that participate in heart development and pathological processes mainly by silencing gene expression. Overwhelming evidence has suggested that miRNAs were involved in various cardiovascular pathological processes, including arrhythmias, ischemia-reperfusion injuries, dysregulation of angiogenesis, mitochondrial abnormalities, fibrosis, and maladaptive remodeling. Various miRNAs could regulate myocardial contractility, vascular proliferation, and mitochondrial function. Meanwhile, it was reported that miRNAs could manipulate nutrition metabolism, especially glucose and lipid metabolism, by regulating insulin signaling pathways, energy substrate transport/metabolism. Recently, increasing studies suggested that the abnormal glucose and lipid metabolism were closely associated with a broad spectrum of cardiovascular diseases (CVDs). Therefore, maintaining glucose and lipid metabolism homeostasis in the heart might be beneficial to CVD patients. In this review, we summarized the present knowledge of the functions of miRNAs in regulating cardiac glucose and lipid metabolism, as well as highlighted the miRNA-based therapies targeting cardiac glucose and lipid metabolism.
Collapse
Affiliation(s)
- Hengzhi Du
- Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yanru Zhao
- Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Huaping Li
- Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
58
|
Rawat PS, Jaiswal A, Khurana A, Bhatti JS, Navik U. Doxorubicin-induced cardiotoxicity: An update on the molecular mechanism and novel therapeutic strategies for effective management. Biomed Pharmacother 2021; 139:111708. [PMID: 34243633 DOI: 10.1016/j.biopha.2021.111708] [Citation(s) in RCA: 412] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/06/2022] Open
Abstract
Doxorubicin (Dox) is a secondary metabolite of the mutated strain of Streptomyces peucetius var. Caesius and belongs to the anthracyclines family. The anti-cancer activity of Dox is mainly exerted through the DNA intercalation and inhibiting topoisomerase II enzyme in fast-proliferating tumors. However, Dox causes cumulative and dose-dependent cardiotoxicity, which results in increased risks of mortality among cancer patients and thus limiting its wide clinical applications. There are several mechanisms has been proposed for doxorubicin-induced cardiotoxicity and oxidative stress, free radical generation and apoptosis are most widely reported. Apart from this, other mechanisms are also involved in Dox-induced cardiotoxicity such as impaired mitochondrial function, a perturbation in iron regulatory protein, disruption of Ca2+ homeostasis, autophagy, the release of nitric oxide and inflammatory mediators and altered gene and protein expression that involved apoptosis. Dox also causes downregulation of DNA methyltransferase 1 (DNMT1) enzyme activity which leads to a reduction in the DNA methylation process. This hypomethylation causes dysregulation in the mitochondrial genes like peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1-alpha (PGC-1α), nuclear respiratory factor 1 (NRF-1) and mitochondrial transcription factor A (TFAM) unit in the heart. Apart from DNA methylation, Dox treatment also alters the micro RNAs levels and histone deacetylase (HDAC) activity. Therefore, in the current review, we have provided a detailed update on the current understanding of the pathological mechanisms behind the well-known Dox-induced cardiotoxicity. Further, we have provided some of the most plausible pharmacological strategies which have been tested against Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Pushkar Singh Rawat
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Aiswarya Jaiswal
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Amit Khurana
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science, PVNRTVU, Rajendranagar, Hyderabad 500030, Telangana, India; Centre for Biomedical Engineering (CBME), Indian Institute of Technology (IIT), Delhi 110016, India.
| | - Jasvinder Singh Bhatti
- Department of human genetics and molecular medicine, School of health sciences, Central University of Punjab, Bathinda 151401, Punjab, India.
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
59
|
miRNA in cardiac development and regeneration. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:14. [PMID: 34060005 PMCID: PMC8166991 DOI: 10.1186/s13619-021-00077-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Ischemic heart disease is one of the main causes of morbidity and mortality in the world. In adult mammalian hearts, most cardiomyocytes are terminally differentiated and have extremely limited capacity of proliferation, making it impossible to regenerate the heart after injuries such as myocardial infarction. MicroRNAs (miRNAs), a class of non-coding single-stranded RNA, which are involved in mRNA silencing and the regulation of post-transcriptional gene expression, have been shown to play a crucial role in cardiac development and cardiomyocyte proliferation. Muscle specific miRNAs such as miR-1 are key regulators of cardiomyocyte maturation and growth, while miR-199-3p and other miRNAs display potent activity to induce proliferation of cardiomyocytes. Given their small size and relative pleiotropic effects, miRNAs have gained significant attraction as promising therapeutic targets or tools in cardiac regeneration. Increasing number of studies demonstrated that overexpression or inhibition of specific miRNAs could induce cardiomyocyte proliferation and cardiac regeneration. Some common targets of pro-proliferation miRNAs, such as the Hippo-Yap signaling pathway, were identified in multiple species, highlighting the power of miRNAs as probes to dissect core regulators of biological processes. A number of miRNAs have been shown to improve heart function after myocardial infarction in mice, and one trial in swine also demonstrated promising outcomes. However, technical difficulties, especially in delivery methods, and adverse effects, such as uncontrolled proliferation, remain. In this review, we summarize the recent progress in miRNA research in cardiac development and regeneration, examine the mechanisms of miRNA regulating cardiomyocyte proliferation, and discuss its potential as a new strategy for cardiac regeneration therapy.
Collapse
|
60
|
Logan SM, Gupta A, Wang A, Levy RJ, Storey KB. Isoflurane and low-level carbon monoxide exposures increase expression of pro-survival miRNA in neonatal mouse heart. Cell Stress Chaperones 2021; 26:541-548. [PMID: 33661504 PMCID: PMC8065082 DOI: 10.1007/s12192-021-01199-0] [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: 01/05/2021] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 11/30/2022] Open
Abstract
Anesthetics such as isoflurane are known to cause apoptosis in the developing mammalian brain. However, isoflurane may have protective effects on the heart via relieving ischemia and downregulating genes related to apoptosis. Ischemic preconditioning, e.g. through the use of low levels of carbon monoxide (CO), has promise in preventing ischemia-reperfusion injury and cell death. However, it is still unclear how it either triggers the stress response in neonatal hearts. For this reason, thirty-three microRNAs (miRNAs) known to be differentially expressed following anesthesia and/or ischemic or hypoxic heart damage were investigated in the hearts from neonatal mice exposed to isoflurane or low level of CO, using an air-exposed control group. Only miR-93-5p increased with isoflurane exposure, which may be associated with the suppression of cell death, autophagy, and inflammation. By contrast, twelve miRNAs were differentially expressed in the heart following CO treatment. Many miRNAs previously shown to be responsible for suppressing cell death, autophagy, and myocardial hypertrophy were upregulated (e.g., 125b-3p, 19-3p, and 21a-5p). Finally, some miRNAs (miR-103-3p, miR-1a-3p, miR-199a-1-5p) which have been implicated in regulating energy balance and cardiac contraction were also differentially expressed. Overall, this study demonstrated that CO-mediated miRNA regulation may promote ischemic preconditioning and cardioprotection based on the putative protective roles of the differentially expressed miRNAs explored herein and the consistency of these results with those that have shown positive effects of CO on heart viability following anesthesia and ischemia-reperfusion stress.
Collapse
Affiliation(s)
- Samantha M Logan
- Institute of Biochemistry & Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Aakriti Gupta
- Institute of Biochemistry & Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Aili Wang
- Department of Anesthesiology, Columbia University Medical Center, 622 West 168th Street, New York, NY, 10032, USA
| | - Richard J Levy
- Department of Anesthesiology, Columbia University Medical Center, 622 West 168th Street, New York, NY, 10032, USA
| | - Kenneth B Storey
- Institute of Biochemistry & Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada.
| |
Collapse
|
61
|
Liu BY, Li L, Liu GL, Ding W, Chang WG, Xu T, Ji XY, Zheng XX, Zhang J, Wang JX. Baicalein attenuates cardiac hypertrophy in mice via suppressing oxidative stress and activating autophagy in cardiomyocytes. Acta Pharmacol Sin 2021; 42:701-714. [PMID: 32796955 PMCID: PMC8115069 DOI: 10.1038/s41401-020-0496-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/29/2020] [Indexed: 12/19/2022]
Abstract
Baicalein is a natural flavonoid extracted from the root of Scutellaria baicalensis that exhibits a variety of pharmacological activities. In this study, we investigated the molecular mechanisms underlying the protective effect of baicalein against cardiac hypertrophy in vivo and in vitro. Cardiac hypertrophy was induced in mice by injection of isoproterenol (ISO, 30 mg·kg-1·d-1) for 15 days. The mice received caudal vein injection of baicalein (25 mg/kg) on 3rd, 6th, 9th, 12th, and 15th days. We showed that baicalein administration significantly attenuated ISO-induced cardiac hypertrophy and restored cardiac function. The protective effect of baicalein against cardiac hypertrophy was also observed in neonatal rat cardiomyocytes treated with ISO (10 μM). In cardiomyocytes, ISO treatment markedly increased reactive oxygen species (ROS) and inhibited autophagy, which were greatly alleviated by pretreatment with baicalein (30 μM). We found that baicalein pretreatment increased the expression of catalase and the mitophagy receptor FUN14 domain containing 1 (FUNDC1) to clear ROS and promote autophagy, thus attenuated ISO-induced cardiac hypertrophy. Furthermore, we revealed that baicalein bound to the transcription factor FOXO3a directly, promoting its transcription activity, and transactivated catalase and FUNDC1. In summary, our data provide new evidence for baicalein and FOXO3a in the regulation of ISO-induced cardiac hypertrophy. Baicalein has great potential for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Bing-Yan Liu
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Ling Li
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Gao-Li Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Wei Ding
- Department of General Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266011, China
| | - Wen-Guang Chang
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Tao Xu
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Xiao-Yu Ji
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Xian-Xin Zheng
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Jing Zhang
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Jian-Xun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China.
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China.
| |
Collapse
|
62
|
Lee Y, Im E. Regulation of miRNAs by Natural Antioxidants in Cardiovascular Diseases: Focus on SIRT1 and eNOS. Antioxidants (Basel) 2021; 10:antiox10030377. [PMID: 33802566 PMCID: PMC8000568 DOI: 10.3390/antiox10030377] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of morbidity and mortality worldwide. The potential benefits of natural antioxidants derived from supplemental nutrients against CVDs are well known. Remarkably, natural antioxidants exert cardioprotective effects by reducing oxidative stress, increasing vasodilation, and normalizing endothelial dysfunction. Recently, considerable evidence has highlighted an important role played by the synergistic interaction between endothelial nitric oxide synthase (eNOS) and sirtuin 1 (SIRT1) in the maintenance of endothelial function. To provide a new perspective on the role of natural antioxidants against CVDs, we focused on microRNAs (miRNAs), which are important posttranscriptional modulators in human diseases. Several miRNAs are regulated via the consumption of natural antioxidants and are related to the regulation of oxidative stress by targeting eNOS and/or SIRT1. In this review, we have discussed the specific molecular regulation of eNOS/SIRT1-related endothelial dysfunction and its contribution to CVD pathologies; furthermore, we selected nine different miRNAs that target the expression of eNOS and SIRT1 in CVDs. Additionally, we have summarized the alteration of miRNA expression and regulation of activities of miRNA through natural antioxidant consumption.
Collapse
Affiliation(s)
| | - Eunok Im
- Correspondence: ; Tel.: +82-51-510-2812; Fax: +82-51-513-6754
| |
Collapse
|
63
|
Song C, Qi H, Liu Y, Chen Y, Shi P, Zhang S, Ren J, Wang L, Cao Y, Sun H. Inhibition of lncRNA Gm15834 Attenuates Autophagy-Mediated Myocardial Hypertrophy via the miR-30b-3p/ULK1 Axis in Mice. Mol Ther 2021; 29:1120-1137. [PMID: 33130312 DOI: 10.1016/j.ymthe.2020.10.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 06/30/2020] [Accepted: 10/23/2020] [Indexed: 12/19/2022] Open
Abstract
Emerging evidence reveals that autophagy plays crucial roles in cardiac hypertrophy. Long noncoding RNAs (lncRNAs) are novel transcripts that function as gene regulators. However, it is unclear whether lncRNAs regulate autophagy in cardiac hypertrophy. Here, we identified a novel transcript named lncRNA Gm15834, which was upregulated in the transverse aortic constriction (TAC) model in vivo and the angiotensin-II (Ang-II)-induced cardiac hypertrophy model in vitro and was regulated by nuclear factor kappa B (NF-κB). Importantly, forced expression of lncRNA Gm15834 enhanced autophagic activity of cardiomyocytes and promoted myocardial hypertrophy, whereas silencing of lncRNA Gm15834 attenuated autophagy-induced myocardial hypertrophy. Mechanistically, we found that lncRNA Gm15834 could function as an endogenous sponge RNA of microRNA (miR)-30b-3p, which was downregulated in cardiac hypertrophy. Inhibition of miR-30b-3p enhanced cardiomyocyte autophagic activity and aggravated myocardial hypertrophy, whereas overexpression of miR-30b-3p suppressed autophagy-induced myocardial hypertrophy by targeting the downstream autophagy factor of unc-51-like kinase 1 (ULK1). Moreover, inhibition of lncRNA Gm15834 by adeno-associated virus carrying short hairpin RNA (shRNA) suppressed cardiomyocyte autophagic activity, improved cardiac function, and mitigated cardiac hypertrophy. Taken together, our study identified a novel regulatory axis encompassing lncRNA Gm15834/miR-30b-3p/ULK1/autophagy in cardiac hypertrophy, which may provide a potential therapy target for cardiac hypertrophy.
Collapse
Affiliation(s)
- Chao Song
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Hanping Qi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Yongsheng Liu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Yunping Chen
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Shu Zhang
- Department of Cardiovascular Medicine, Fifth Clinical College of Harbin Medical University, Daqing, Heilongjiang 163316, China
| | - Jing Ren
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Lixin Wang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China.
| |
Collapse
|
64
|
Lu J, Liu Y, Wang W, Li P, Qi F. Knockdown of miR-146a in regulatory T cells suppresses heart transplantation rejection in mice by increasing autophagy. Transpl Immunol 2021; 65:101372. [PMID: 33581286 DOI: 10.1016/j.trim.2021.101372] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 01/10/2023]
Abstract
Clinical trials of regulatory T cells (Tregs) have shown that adoptive transfer of Tregs has great promise for the treatment of rejection. However, strategies to improve Treg function are needed in order to enhance their efficacy and reduce the number of Tregs required for adoptive transfer. Autophagy is a process for degrading intracellular components, and it mediates cell death, lymphocyte homeostasis, and Treg function. Studies have shown that the survival and function of Tregs with disrupted autophagy are defective. We found that the autophagic status of Tregs was compromised during acute rejection, allowing us to enhance Treg autophagy by regulating microRNA-146a (miR-146a), which is highly expressed in Tregs and is implicated in their function and metabolism. MiR-146a antagomir-mediated miR-146a knockdown promoted Treg autophagy, as evaluated by Western blot analysis. Further, we evaluated whether altering autophagy affects Treg function in both an in vitro cell coculture model and a heart transplantation model in mice. An increase in autophagy enhanced the inhibitory effects of Tregs on CD4+ T cells and dendritic cells (DCs) in vivo and in vitro. In addition, adoptive transfer of highly autophagic Tregs treated with miR-146a antagomir significantly alleviated rejection. Collectively, these data provide a new method that uses miR-146a knockdown to increase Treg efficacy by increasing autophagy.
Collapse
Affiliation(s)
- Jian Lu
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China
| | - Yanhong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China
| | - Weiwei Wang
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China; Department of General Surgery, Tianjin Medical University Baodi Clinical College, No. 8, Guangchuan Road, Baodi District, Tianjin 301800, China
| | - Peiyuan Li
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China
| | - Feng Qi
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China.
| |
Collapse
|
65
|
Liu B, Wang B, Zhang X, Lock R, Nash T, Vunjak-Novakovic G. Cell type-specific microRNA therapies for myocardial infarction. Sci Transl Med 2021; 13:eabd0914. [PMID: 33568517 PMCID: PMC8848299 DOI: 10.1126/scitranslmed.abd0914] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 01/19/2021] [Indexed: 12/13/2022]
Abstract
Current interventions fail to recover injured myocardium after infarction and prompt the need for development of cardioprotective strategies. Of increasing interest is the therapeutic use of microRNAs to control gene expression through specific targeting of mRNAs. In this Review, we discuss current microRNA-based therapeutic strategies, describing the outcomes and limitations of key microRNAs with a focus on target cell types and molecular pathways. Last, we offer a perspective on the outlook of microRNA therapies for myocardial infarction, highlighting the outstanding challenges and emerging strategies.
Collapse
Affiliation(s)
- Bohao Liu
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Bryan Wang
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Xiaokan Zhang
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Roberta Lock
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Trevor Nash
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Gordana Vunjak-Novakovic
- Department of Medicine, Columbia University, New York, NY 10032, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| |
Collapse
|
66
|
Sciarretta S, Forte M, Frati G, Sadoshima J. The complex network of mTOR signaling in the heart. Cardiovasc Res 2021; 118:424-439. [PMID: 33512477 DOI: 10.1093/cvr/cvab033] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/13/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) integrates several intracellular and extracellular signals involved in the regulation of anabolic and catabolic processes. mTOR assembles into two macromolecular complexes, named mTORC1 and mTORC2, which have different regulators, substrates and functions. Studies of gain- and loss-of-function animal models of mTOR signaling revealed that mTORC1/2 elicit both adaptive and maladaptive functions in the cardiovascular system. Both mTORC1 and mTORC2 are indispensable for driving cardiac development and cardiac adaption to stress, such as pressure overload. However, persistent and deregulated mTORC1 activation in the heart is detrimental during stress and contributes to the development and progression of cardiac remodeling and genetic and metabolic cardiomyopathies. In this review, we discuss the latest findings regarding the role of mTOR in the cardiovascular system, both under basal conditions and during stress, such as pressure overload, ischemia and metabolic stress. Current data suggest that mTOR modulation may represent a potential therapeutic strategy for the treatment of cardiac diseases.
Collapse
Affiliation(s)
- Sebastiano Sciarretta
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,IRCCS Neuromed, Pozzilli (IS), Italy
| | | | - Giacomo Frati
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,IRCCS Neuromed, Pozzilli (IS), Italy
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
67
|
Riching AS, Song K. Cardiac Regeneration: New Insights Into the Frontier of Ischemic Heart Failure Therapy. Front Bioeng Biotechnol 2021; 8:637538. [PMID: 33585427 PMCID: PMC7873479 DOI: 10.3389/fbioe.2020.637538] [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: 12/03/2020] [Accepted: 12/29/2020] [Indexed: 12/17/2022] Open
Abstract
Ischemic heart disease is the leading cause of morbidity and mortality in the world. While pharmacological and surgical interventions developed in the late twentieth century drastically improved patient outcomes, mortality rates over the last two decades have begun to plateau. Following ischemic injury, pathological remodeling leads to cardiomyocyte loss and fibrosis leading to impaired heart function. Cardiomyocyte turnover rate in the adult heart is limited, and no clinical therapies currently exist to regenerate cardiomyocytes lost following ischemic injury. In this review, we summarize the progress of therapeutic strategies including revascularization and cell-based interventions to regenerate the heart: transiently inducing cardiomyocyte proliferation and direct reprogramming of fibroblasts into cardiomyocytes. Moreover, we highlight recent mechanistic insights governing these strategies to promote heart regeneration and identify current challenges in translating these approaches to human patients.
Collapse
Affiliation(s)
- Andrew S. Riching
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kunhua Song
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
68
|
Ye H, Xu G, Zhang D, Wang R. The protective effects of the miR-129-5p/keap-1/Nrf2 axis on Ang II-induced cardiomyocyte hypertrophy. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:154. [PMID: 33569456 PMCID: PMC7867905 DOI: 10.21037/atm-20-8079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background Cardiac hypertrophy is a common pathological process in many cardiac diseases, and persistent cardiac hypertrophy is the main cause of heart failure and sudden cardiogenic death. Thus, it is essential to elucidate the mechanism of cardiac hypertrophy to ensure better prevention and treatment. Methods The Human cardiac myocytes (HCMs) were incubated with 100 nmol/L Ang II (Sigma) for 48 hours to induce the in vitro cardiomyocyte hypertrophy model. The [(3H])-leucine incorporation assay was used to evaluate cardiomyocytes hypertrophy. The activities of oxidative stress related enzymes superoxide dismutase (SOD), catalase (CAT), malondialdehyde (MDA) and nitric oxide (NO) were detected using corresponding detection kits following standard protocol. Targeting relationship was verified through Bioinformatics analysis and luciferase reporter gene assay. The morphological change of cardiomyocyte was observed through immunofluorescence staining. Expressions of message ribonucleic acid (mRNA) and proteins were detected by quantitative real-time polymerase chain reaction and western blot, respectively. Results In our study, the suppressed expression of micro ribonucleic acid (miRNA)-129-5p and the elevated expression of kelch-like ECH-associated protein 1 (keap-1) were found in the angiotensin II (Ang II)-induced cardiomyocyte hypertrophy model. MiR-129-5p effectively mimics suppressed Ang II-induced hypertrophic responses and oxidative stress. The results also showed that keap-1 was a target of miR-129-5p, and that the miR-129-5p inhibitor promoted cardiomyocyte hypertrophy and oxidative stress by elevating keap-1. Additionally, small interfering RNA (siRNA)-keap-1 activated the nuclear factor erythroid2-related factor 2 (Nrf2) pathway, while the miR-129-5p inhibitor inactivated the Nrf2 pathway by further elevating keap-1. The addition of the Nrf2 pathway activator NK-252 largely weakened the promoting effects of the miR-129-5p inhibitor on the progression of cardiomyocyte hypertrophy by suppressing oxidative stress. Conclusions In general, the results indicate that the overexpression of miRNA-129-5p protects against cardiomyocyte hypertrophy by targeting keap-1 via the Nrf2 pathway.
Collapse
Affiliation(s)
- Huiming Ye
- Department of Cardiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Guiyu Xu
- Department of Cardiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Dexian Zhang
- Department of Cardiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Rupeng Wang
- Department of Cardiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
69
|
Affiliation(s)
- Ajit Magadum
- Cardiovascular Research Center (A.M., A.A.K., E.C., Y.S., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Department of Genetics and Genomic Sciences (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Black Family Stem Cell Institute (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York
| | - Ann Anu Kurian
- Cardiovascular Research Center (A.M., A.A.K., E.C., Y.S., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Department of Genetics and Genomic Sciences (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Black Family Stem Cell Institute (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York
| | - Elena Chepurko
- Cardiovascular Research Center (A.M., A.A.K., E.C., Y.S., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Department of Genetics and Genomic Sciences (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Black Family Stem Cell Institute (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York
| | - Yassine Sassi
- Cardiovascular Research Center (A.M., A.A.K., E.C., Y.S., L.Z.), Icahn School of Medicine at Mount Sinai, New York
| | - Roger J Hajjar
- Phospholamban Foundation, Amsterdam, The Netherlands (R.J.H.)
| | - Lior Zangi
- Cardiovascular Research Center (A.M., A.A.K., E.C., Y.S., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Department of Genetics and Genomic Sciences (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Black Family Stem Cell Institute (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York
| |
Collapse
|
70
|
Li Y, Luan Y, Li J, Song H, Li Y, Qi H, Sun B, Zhang P, Wu X, Liu X, Yang Y, Tao W, Cai L, Yang Z, Yang Y. Exosomal miR-199a-5p promotes hepatic lipid accumulation by modulating MST1 expression and fatty acid metabolism. Hepatol Int 2020; 14:1057-1074. [PMID: 33037981 DOI: 10.1007/s12072-020-10096-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/12/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Non-alcoholic fatty liver disease (NAFLD) and its complications has become an expanding health problem worldwide with limited therapeutic approaches. The current study was aiming to identify novel microRNA in the regulation of hepatic lipid metabolism in NAFLD. APPROCHES AND RESULTS Systematic screening of microRNA expression by high-throughput small RNA sequencing demonstrated that microRNA 199a-5p (miR-199a-5p) was significantly upregulated in high fat diet-induced steatosis mouse model, with the most abundant expression in adipose tissue. MST1 was further identified as the target gene for miR-199a with specific recognition at the 3' untranslated region with dural luciferase reporter assay. Delivery of miR-199a-5p with exosomes into mice aggravated liver lipid accumulation in hepatocytes, accompanied by down-regulation of hepatic MST1 expression and modulation of hepatic lipogenesis and lipolysis, including SREBP-1c, AMPK signaling cascades and the down-stream CPT1α and FASN. Conversely, administration of exosome containing anti-miR-199a-5p resulted in attenuated steotosis in mice fed on high fat diet. Importanly, miR-199a-5p-induced abnormal cellular lipid accumulation could be markedly reversed by overexpression of MST1. CONCLUSION miR-199a-5p might be an essentail regulator for hepatic lipid metabolism, possibly through its interction with MST1 and the subsequent signaling cascade. Thus, miR-199a-5p may serve as an important therapeutic target in the treatment of NAFLD.
Collapse
Affiliation(s)
- Yuhan Li
- School of Basic Medical Sciences, Ningxia Medical University, Xingqing District, 692 Shengli St, Yinchuan, 75004, China
- The Institute of Endocrinology, Ningxia Medical University, Yinchuan, China
- Institute of Laboratory Animal Science (ILAS), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), #5 Pan Jia Yuan Nan Li, Chaoyang Districe, Beijing, 100021, China
| | - Yansong Luan
- School of Basic Medical Sciences, Ningxia Medical University, Xingqing District, 692 Shengli St, Yinchuan, 75004, China
- The Institute of Endocrinology, Ningxia Medical University, Yinchuan, China
| | - Jianning Li
- School of Basic Medical Sciences, Ningxia Medical University, Xingqing District, 692 Shengli St, Yinchuan, 75004, China
- The Institute of Endocrinology, Ningxia Medical University, Yinchuan, China
| | - Hui Song
- School of Basic Medical Sciences, Ningxia Medical University, Xingqing District, 692 Shengli St, Yinchuan, 75004, China
- The Institute of Endocrinology, Ningxia Medical University, Yinchuan, China
| | - Yan Li
- School of Basic Medical Sciences, Ningxia Medical University, Xingqing District, 692 Shengli St, Yinchuan, 75004, China
- The Institute of Endocrinology, Ningxia Medical University, Yinchuan, China
| | - Hi Qi
- School of Basic Medical Sciences, Ningxia Medical University, Xingqing District, 692 Shengli St, Yinchuan, 75004, China
| | - Bo Sun
- Second Affiliated Hospital, Ningxia Medical University, Yinchuan, China
| | - Peng Zhang
- Second Affiliated Hospital, Ningxia Medical University, Yinchuan, China
| | - Xianxian Wu
- Institute of Laboratory Animal Science (ILAS), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), #5 Pan Jia Yuan Nan Li, Chaoyang Districe, Beijing, 100021, China
| | - Xing Liu
- Institute of Laboratory Animal Science (ILAS), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), #5 Pan Jia Yuan Nan Li, Chaoyang Districe, Beijing, 100021, China
| | - Yanhui Yang
- School of Basic Medical Sciences, Ningxia Medical University, Xingqing District, 692 Shengli St, Yinchuan, 75004, China
- The Institute of Endocrinology, Ningxia Medical University, Yinchuan, China
| | - Wufan Tao
- Institute of Developmental Biology and Molecular Medicine and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Lei Cai
- Department of Physiology, Cardiovascular Research Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Zhiwei Yang
- Institute of Laboratory Animal Science (ILAS), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), #5 Pan Jia Yuan Nan Li, Chaoyang Districe, Beijing, 100021, China.
| | - Yi Yang
- School of Basic Medical Sciences, Ningxia Medical University, Xingqing District, 692 Shengli St, Yinchuan, 75004, China.
- The Institute of Endocrinology, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
71
|
Tang L, Xie J, Yu X, Zheng Y. MiR-26a-5p inhibits GSK3β expression and promotes cardiac hypertrophy in vitro. PeerJ 2020; 8:e10371. [PMID: 33240671 PMCID: PMC7678492 DOI: 10.7717/peerj.10371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 10/26/2020] [Indexed: 12/03/2022] Open
Abstract
Background The role of miR-26a-5p expression in cardiac hypertrophy remains unclear. Herein, the effect of miR-26a-5p on cardiac hypertrophy was investigated using phenylephrine (PE)-induced cardiac hypertrophy in vitro and in a rat model of hypertension-induced hypertrophy in vivo. Methods The PE-induced cardiac hypertrophy models in vitro and vivo were established. To investigate the effect of miR-26a-5p activation on autophagy, the protein expression of autophagosome marker (LC3) and p62 was detected by western blot analysis. To explore the effect of miR-26a-5p activation on cardiac hypertrophy, the relative mRNA expression of cardiac hypertrophy related mark GSK3β was detected by qRT-PCR in vitro and vivo. In addition, immunofluorescence staining was used to detect cardiac hypertrophy related mark α-actinin. The cell surface area was measured by immunofluorescence staining. The direct target relationship between miR-26a-5p and GSK3β was confirmed by dual luciferase report. Results MiR-26a-5p was highly expressed in PE-induced cardiac hypertrophy. MiR-26a-5p promoted LC3II and decreased p62 expression in PE-induced cardiac hypertrophy in the presence or absence of lysosomal inhibitor. Furthermore, miR-26a-5p significantly inhibited GSK3β expression in vitro and in vivo. Dual luciferase report results confirmed that miR-26a-5p could directly target GSK3β. GSK3β overexpression significantly reversed the expression of cardiac hypertrophy-related markers including ANP, ACTA1 and MYH7. Immunofluorescence staining results demonstrated that miR-26a-5p promoted cardiac hypertrophy related protein α-actinin expression, and increased cell surface area in vitro and in vivo. Conclusion Our study revealed that miR-26a-5p promotes myocardial cell autophagy activation and cardiac hypertrophy by regulating GSK3β, which needs further research.
Collapse
Affiliation(s)
- Liqun Tang
- Department of Geriatrics, Zhejiang Province People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jianhong Xie
- Department of Geriatrics, Zhejiang Province People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoqin Yu
- Department of Geriatrics, Zhejiang Aid Hospital, Hangzhou, Zhejiang, China
| | - Yangyang Zheng
- Department of Geriatrics, Zhejiang Province People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
72
|
Yan H, Wang H, Zhu X, Huang J, Li Y, Zhou K, Hua Y, Yan F, Wang DZ, Luo Y. Adeno-associated virus-mediated delivery of anti-miR-199a tough decoys attenuates cardiac hypertrophy by targeting PGC-1alpha. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 23:406-417. [PMID: 33473326 PMCID: PMC7787996 DOI: 10.1016/j.omtn.2020.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/10/2020] [Indexed: 02/08/2023]
Abstract
MicroRNAs (miRNAs) are important regulators in the process of cardiac hypertrophy and heart failure. Previous studies have shown that miR-199a is upregulated in pressure-overload cardiac hypertrophy and that inhibition of miR-199a attenuates cardiac hypertrophy in vitro. However, the therapeutic role of anti-miR-199a treatment in the cardiac hypertrophy in vivo model is less known. Here, we show an efficient and useful method to treat mouse cardiac hypertrophy and restore cardiac function through injection of adeno-associated virus (AAV)-mediated anti-miR-199a tough decoys (TuDs). RNA-seq transcriptome analysis indicated that genes related to cytoplasmic translation and mitochondrial respiratory chain complex assembly were upregulated in anti-miR-199a-treated recovered hearts. We further validated that PGC-1α is the direct target of miR-199a involved in the therapeutic effect and the regulation of the PGC-1α/ERRα axis and that the downstream pathway of mitochondrial fatty acid oxidation and oxidative phosphorylation constitute the underlying mechanism of the restored mitochondrial structure and function in our anti-miR-199a-treated mice. Our study highlights the important regulatory role of miR-199a in cardiac hypertrophy and the value of the AAV-mediated miRNA delivery system.
Collapse
Affiliation(s)
- Hualin Yan
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Wang
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoxia Zhu
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianbo Huang
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yifei Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Kaiyu Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yimin Hua
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Yan
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Yan Luo
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
73
|
Gao J, Chen X, Shan C, Wang Y, Li P, Shao K. Autophagy in cardiovascular diseases: role of noncoding RNAs. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 23:101-118. [PMID: 33335796 PMCID: PMC7732971 DOI: 10.1016/j.omtn.2020.10.039] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular diseases (CVDs) remain the world's leading cause of death. Cardiomyocyte autophagy helps maintain normal metabolism and functioning of the heart. Importantly, mounting evidence has revealed that autophagy plays a dual role in CVD pathology. Under physiological conditions, moderate autophagy maintains cell metabolic balance by degrading and recycling damaged organelles and proteins, and it promotes myocardial survival, but excessive or insufficient autophagy is equally deleterious and contributes to disease progression. Noncoding RNAs (ncRNAs) are a class of RNAs transcribed from the genome, but most ncRNAs do not code for functional proteins. In recent years, increasingly, various ncRNAs have been identified, and they play important regulatory roles in the physiological and pathological processes of organisms, as well as in autophagy. Thus, determining whether ncRNA-regulated autophagy plays a protective role in CVDs or promotes their progression can help us to develop ncRNAs as therapeutic targets in autophagy-related CVDs. In this review, we briefly summarize the regulatory roles of several important ncRNAs, including microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), in the autophagy of various CVDs to provide a theoretical basis for the etiology and pathogenesis of CVDs and develop novel therapies to treat CVDs.
Collapse
Affiliation(s)
- Jinning Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Xiatian Chen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Chan Shan
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Kai Shao
- Department of Central Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, China
| |
Collapse
|
74
|
Non-coding RNAs: The key detectors and regulators in cardiovascular disease. Genomics 2020; 113:1233-1246. [PMID: 33164830 DOI: 10.1016/j.ygeno.2020.10.024] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/27/2020] [Accepted: 10/20/2020] [Indexed: 12/22/2022]
Abstract
Cardiovascular disease (CVD) is an important cause of disease-related death worldwide. One of its main pathological bases is imbalances in gene expression. Non-coding RNAs are a class of transcripts that do not encode proteins. They include microRNA (miRNA), long noncoding RNA (lncRNA) and circular RNA (circRNA). They have important biological functions such as regulating transcription and translation, as well as interacting with DNA, RNA, and proteins. They are also closely associated with pathological processes in CVD. This review will focus on the expression and function of miRNA, lncRNA, circRNA, as well as on their roles and molecular mechanisms in CVDs such as cardiac hypertrophy, heart failure, arrhythmia, myocardial infarction, atherosclerosis, rheumatic heart disease, myocardial fibrosis, pulmonary arterial hypertension. This review will outline concepts provide bases for early diagnosis and targeted treatment of CVDs.
Collapse
|
75
|
Zhang L, Ding H, Zhang Y, Wang Y, Zhu W, Li P. Circulating MicroRNAs: Biogenesis and Clinical Significance in Acute Myocardial Infarction. Front Physiol 2020; 11:1088. [PMID: 33013463 PMCID: PMC7494963 DOI: 10.3389/fphys.2020.01088] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
Acute myocardial infarction (AMI) causes many deaths around the world. Early diagnosis can prevent the development of AMI and provide theoretical support for the subsequent treatment. miRNAs participate in the AMI pathological processes. We aim to determine the early diagnostic and the prognostic roles of circulating miRNAs in AMI in the existing studies and summarize all the data to provide a greater understanding of their utility for clinical application. We reviewed current knowledge focused on the AMI development and circulating miRNA formation. Meanwhile, we collected and analyzed the potential roles of circulating miRNAs in AMI diagnosis, prognosis and therapeutic strategies. Additionally, we elaborated on the challenges and clinical perspectives of the application of circulating miRNAs in AMI diagnosis. Circulating miRNAs are stable in the circulation and have earlier increases of circulating levels than diagnostic golden criteria. In addition, they are tissue and disease-specific. All these characteristics indicate that circulating miRNAs are promising biomarkers for the early diagnosis of AMI. Although there are several limitations to be resolved before clinical use, the application of circulating miRNAs shows great potential in the early diagnosis and the prognosis of AMI.
Collapse
Affiliation(s)
- Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Han Ding
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Wenjie Zhu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
76
|
Liu JJ, Tang MM, Zhu ML, Xie CX, Kang PF, Ling X, Zhang H, Wang XJ, Tang B. Docosahexaenoic acid inhibits Ca 2+ influx and downregulates CaSR by upregulating microRNA-16 in pulmonary artery smooth muscle cells. J Biochem Mol Toxicol 2020; 34:e22573. [PMID: 32659049 DOI: 10.1002/jbt.22573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/25/2020] [Accepted: 06/23/2020] [Indexed: 11/10/2022]
Abstract
Docosahexaenoic acid (DHA) is reported to have the potential to ameliorate pulmonary arterial hypertension (PAH), while the specific mechanism is still obscure. This study aims to investigate the function of DHA in pulmonary artery smooth muscle cells (PASMCs) and explore the underlying mechanism. In our study, DHA was used to incubate PASMCs. Cytosolic-free Ca2+ concentration ([Ca2+ ]cyt) was measured using Fluo-3 AM method. Real-time polymerase chain reaction was used to detect microRNA-16 (miR-16) and calcium-sensing receptor (CaSR) messenger RNA expression levels. CCK-8 assay, BrdU assay, and Transwell assay were employed to detect the effects of DHA on proliferation and migration of PASMCs. CaSR was confirmed as a direct target of miR-16 using dual-luciferase assay, polymerase chain reaction, and Western blot analysis. It was found that DHA significantly inhibited PASMC proliferation and migration and decreased [Ca2+ ]cyt. After transfection of miR-16 mimics, proliferation and migration ability of PASMCs were significantly inhibited, whereas opposite effects were observed after miR-16 inhibition. [Ca2+ ]cyt was also inhibited by miR-16 transfection. DHA then promoted the expression of miR-16, and the effects of DHA on PASMCs were annulled when miR-16 was inhibited. CaSR was identified as a direct target of miR-16. CaSR was inhibited directly by miR-16 and indirectly by DHA. In conclusion, DHA inhibits the proliferation and migration of PASMCs, and probably ameliorates PAH via regulating miR-16/CaSR axis.
Collapse
Affiliation(s)
- Jin-Jun Liu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Ming-Ming Tang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Ming-Li Zhu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Cai-Xia Xie
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Pin-Fang Kang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Xuan Ling
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Heng Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Xiao-Jing Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Bi Tang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| |
Collapse
|
77
|
Promoting role of circ-Jarid2/miR-129-5p/Celf1 axis in cardiac hypertrophy. Gene Ther 2020; 28:718-728. [PMID: 32632266 DOI: 10.1038/s41434-020-0165-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 05/20/2020] [Accepted: 06/02/2020] [Indexed: 11/09/2022]
Abstract
Cardiac hypertrophy is imposed much pressure on heart and threatening our live. Previous study suggested that dysregulation of Celf1 is largely connecting to neonatal cardiac dysfunction. Hence, we aimed to explore the precise function and probable regulatory mechanism upstream of Celf1in cardiac hypertrophy. Here, Ang-II treatment was implemented to stimulate hypertrophic phenotypes inH9C2 and MCM cells. Immunofluorescence assay was conducted to measure the surface area of cardiomyocytes. And qRT-PCR assay was conducted to investigate gene expression. Moreover, western blot assay was conducted to probe the protein levels. Results uncovered that Celf1 expression was increased dependent on elevated Ang-II concentration, and that inhibited Celf1 could relieve the Ang-II-caused cardiac hypertrophy. Significantly, Celf1was found to be targeted by miR-129-5p but then released via the sponging role of circ-Jarid2. Furthermore, circ-Jarid2 was found to promote cardiac hypertrophy, whereas miR-129-5p played suppressing parts in hypertrophic cardiomyocytes. Moreover, we verified circ-Jarid2 contributed to cardiac hypertrophy via miR-129-5p/Celf1 axis both in vitro and in vivo. In conclusion, circ-Jarid2/miR-129-5p/Celf1 axis aggravates cardiac hypertrophy, which provides new ideas for developing treatment strategies for patients with cardiac hypertrophy.
Collapse
|
78
|
Talebian S, Daghagh H, Yousefi B, Ȍzkul Y, Ilkhani K, Seif F, Alivand MR. The role of epigenetics and non-coding RNAs in autophagy: A new perspective for thorough understanding. Mech Ageing Dev 2020; 190:111309. [PMID: 32634442 DOI: 10.1016/j.mad.2020.111309] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/22/2020] [Accepted: 06/28/2020] [Indexed: 12/18/2022]
Abstract
Autophagy is a major self-degradative intracellular process required for the maintenance of homeostasis and promotion of survival in response to starvation. It plays critical roles in a large variety of physiological and pathological processes. On the other hand, aberrant regulation of autophagy can lead to various cancers and neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Crohn's disease. Emerging evidence strongly supports that epigenetic signatures, related non-coding RNA profiles, and their cross-talking are significantly associated with the control of autophagic responses. Therefore, it may be helpful and promising to manage autophagic processes by finding valuable markers and therapeutic approaches. Although there is a great deal of information on the components of autophagy in the cytoplasm, the molecular basis of the epigenetic regulation of autophagy has not been completely elucidated. In this review, we highlight recent research on epigenetic changes through the expression of autophagy-related genes (ATGs), which regulate autophagy, DNA methylation, histone modifications as well as non-coding RNAs, including long non-coding RNAs (lncRNAs), microRNAs (miRNAs) and their relationship with human diseases, that play key roles in causing autophagy-related diseases.
Collapse
Affiliation(s)
- Shahrzad Talebian
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Daghagh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yusuf Ȍzkul
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Khandan Ilkhani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Seif
- Department of Immunology & Allergy, Academic Center for Education, Culture, and Research, Tehran, Iran
| | - Mohammad Reza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
79
|
Wang L, Wang J, Li G, Xiao J. Non-coding RNAs in Physiological Cardiac Hypertrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:149-161. [PMID: 32285410 DOI: 10.1007/978-981-15-1671-9_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Non-coding RNA (ncRNA) is a class of RNAs that are not act as translational protein templates. They are involved in the regulation of gene transcription, RNA maturation and protein translation, participating in a variety of physiological and physiological processes. NcRNAs have important functions, and are recently one of the hotspots in biomedical research. Cardiac hypertrophy is classified into physiological cardiac hypertrophy and pathological cardiac hypertrophy. Different from pathological cardiac hypertrophy, physiological cardiac hypertrophy usually developed during exercise, pregnancy, normal postnatal growth, accompanied with preservation or improvement of systolic function, while no cardiac fibrosis. In this chapter, we will briefly introduce the definition, characteristics, and functions of ncRNAs, including miRNAs, lncRNAs, and circRNAs, as well as a summary of the existing bioinformatics online databases which commonly used in the study of ncRNAs. Specially, this chapter will be focused on the characteristics and the underlying mechanisms about physiological cardiac hypertrophy. Furthermore, the regulatory mechanism of ncRNAs in physiological hypertrophy and the latest research progress will be summarized. Taken together, exploring physiologic cardiac hypertrophy-specific ncRNAs might be a unique research perspective that provides new point of view for interventions in heart failure and other cardiovascular diseases.
Collapse
Affiliation(s)
- Lijun Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Jiaqi Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China.
| |
Collapse
|
80
|
Xu CN, Kong LH, Ding P, Liu Y, Fan ZG, Gao EH, Yang J, Yang LF. Melatonin ameliorates pressure overload-induced cardiac hypertrophy by attenuating Atg5-dependent autophagy and activating the Akt/mTOR pathway. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165848. [PMID: 32473999 DOI: 10.1016/j.bbadis.2020.165848] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/06/2020] [Accepted: 05/21/2020] [Indexed: 12/16/2022]
Abstract
Cardiac hypertrophy, including hypertension and valvular dysfunction, is a pathological feature of many cardiac diseases that ultimately leads to heart failure. Melatonin confers a protective role against pathological cardiac hypertrophy, but the underlying mechanisms remain elusive. In the present study, we hypothesized that melatonin protects against pressure overload-induced cardiac hypertrophy by attenuating Atg5-dependent autophagy and activating the Akt/mTOR pathway. Male C57BL/6 mice that received adenovirus carrying cardiac-specific Atg5 (under the cTNT promoter; Ad-cTNT-Atg5) underwent transverse aortic constriction (TAC) or sham operation and received an intraperitoneal injection of melatonin (10 mg/kg/d), vehicle or LY294002 (10 mg/kg/d) for 8 weeks. Melatonin treatment for 8 weeks markedly attenuated cardiac hypertrophy and restored impaired cardiac function, as indicated by a decreased HW/BW ratio, reduced cell cross-sectional area and fibrosis, downregulated the mRNA levels of ANP, BNP, and β-MHC and ameliorated adverse effects on the LVEF and LVFS. Melatonin treatment also inhibited apoptosis and alleviated autophagy dysfunction. Furthermore, melatonin inhibited Akt/mTOR pathway activation, while these effects were blocked by LY294002. In addition, the effect of melatonin regulation on TAC-induced autophagy dysfunction was inhibited by LY294002 or cardiac-specific Atg5 overexpression. As expected, Akt/mTOR pathway inhibition or cardiac-specific Atg5 overexpression restrained melatonin alleviation of pressure overload-induced cardiac hypertrophy. These results demonstrated that melatonin ameliorated pressure overload-induced cardiac hypertrophy by attenuating Atg5-dependent autophagy and activating the Akt/mTOR pathway.
Collapse
Affiliation(s)
- Chen-Nian Xu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Ling-Heng Kong
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China; School of Basic Medical Science, Xi'an Medical University, Xi'an 710021, China
| | - Peng Ding
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Yang Liu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Zhen-Ge Fan
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Er-He Gao
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jian Yang
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, China.
| | - Li-Fang Yang
- Department of Anesthesiology, Xi'an Children's Hospital, Xi'an 710003, China.
| |
Collapse
|
81
|
Yun HR, Jo YH, Kim J, Shin Y, Kim SS, Choi TG. Roles of Autophagy in Oxidative Stress. Int J Mol Sci 2020; 21:ijms21093289. [PMID: 32384691 PMCID: PMC7246723 DOI: 10.3390/ijms21093289] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a catabolic process for unnecessary or dysfunctional cytoplasmic contents by lysosomal degradation pathways. Autophagy is implicated in various biological processes such as programmed cell death, stress responses, elimination of damaged organelles and development. The role of autophagy as a crucial mediator has been clarified and expanded in the pathological response to redox signalling. Autophagy is a major sensor of the redox signalling. Reactive oxygen species (ROS) are highly reactive molecules that are generated as by-products of cellular metabolism, principally by mitochondria. Mitochondrial ROS (mROS) are beneficial or detrimental to cells depending on their concentration and location. mROS function as redox messengers in intracellular signalling at physiologically low level, whereas excessive production of mROS causes oxidative damage to cellular constituents and thus incurs cell death. Hence, the balance of autophagy-related stress adaptation and cell death is important to comprehend redox signalling-related pathogenesis. In this review, we attempt to provide an overview the basic mechanism and function of autophagy in the context of response to oxidative stress and redox signalling in pathology.
Collapse
Affiliation(s)
- Hyeong Rok Yun
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
| | - Yong Hwa Jo
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Jieun Kim
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (S.S.K.); (T.G.C.); Tel.: +82-2-961-0524 (S.S.K.); +82-2-961-0287 (T.G.C.)
| | - Tae Gyu Choi
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (S.S.K.); (T.G.C.); Tel.: +82-2-961-0524 (S.S.K.); +82-2-961-0287 (T.G.C.)
| |
Collapse
|
82
|
Abstract
Cardiac hypertrophy is a significant risk factor for cardiovascular disease, including heart failure, arrhythmia, and sudden death. Cardiac hypertrophy involves both embryonic gene expression and transcriptional reprogramming, which are tightly regulated by epigenetic mechanisms. An increasing number of studies have demonstrated that epigenetics plays an influential role in the occurrence and development of cardiac hypertrophy. Here, we summarize the latest research progress on epigenetics in cardiac hypertrophy involving DNA methylation, histone modification, and non-coding RNA, to help understand the mechanism of epigenetics in cardiac hypertrophy. The expression of both embryonic and functional genes can be precisely regulated by epigenetic mechanisms during cardiac hypertrophy, providing a substantial number of therapeutic targets. Thus, epigenetic treatment is expected to become a novel therapeutic strategy for cardiac hypertrophy. According to the research performed to date, epigenetic mechanisms associated with cardiac hypertrophy remain far from completely understood. Therefore, epigenetic mechanisms require further exploration to improve the prevention, diagnosis, and treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Hao Lei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Kaijun Sun
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
83
|
Protein and Mitochondria Quality Control Mechanisms and Cardiac Aging. Cells 2020; 9:cells9040933. [PMID: 32290135 PMCID: PMC7226975 DOI: 10.3390/cells9040933] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease (CVD) is the number one cause of death in the United States. Advancing age is a primary risk factor for developing CVD. Estimates indicate that 20% of the US population will be ≥65 years old by 2030. Direct expenditures for treating CVD in the older population combined with indirect costs, secondary to lost wages, are predicted to reach $1.1 trillion by 2035. Therefore, there is an eminent need to discover novel therapeutic targets and identify new interventions to delay, lessen the severity, or prevent cardiovascular complications associated with advanced age. Protein and organelle quality control pathways including autophagy/lysosomal and the ubiquitin-proteasome systems, are emerging contributors of age-associated myocardial dysfunction. In general, two findings have sparked this interest. First, strong evidence indicates that cardiac protein degradation pathways are altered in the heart with aging. Second, it is well accepted that damaged and misfolded protein aggregates and dysfunctional mitochondria accumulate in the heart with age. In this review, we will: (i) define the different protein and mitochondria quality control mechanisms in the heart; (ii) provide evidence that each quality control pathway becomes dysfunctional during cardiac aging; and (iii) discuss current advances in targeting these pathways to maintain cardiac function with age.
Collapse
|
84
|
Fulgencio-Covián A, Alonso-Barroso E, Guenzel AJ, Rivera-Barahona A, Ugarte M, Pérez B, Barry MA, Pérez-Cerdá C, Richard E, Desviat LR. Pathogenic implications of dysregulated miRNAs in propionic acidemia related cardiomyopathy. Transl Res 2020; 218:43-56. [PMID: 31951825 DOI: 10.1016/j.trsl.2019.12.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/03/2019] [Accepted: 12/23/2019] [Indexed: 12/25/2022]
Abstract
Cardiac alterations (hypertrophic/dilated cardiomyopathy, and rhythm alterations) are one of the major causes of mortality and morbidity in propionic acidemia (PA), caused by the deficiency of the mitochondrial enzyme propionyl-CoA carboxylase (PCC), involved in the catabolism of branched-chain amino acids, cholesterol, and odd-chain fatty acids. Impaired mitochondrial oxidative phosphorylation has been documented in heart biopsies of PA patients, as well as in the hypomorphic Pcca-/-(A138T) mouse model, in the latter correlating with increased oxidative damage and elevated expression of cardiac dysfunction biomarkers atrial and brain natriuretic peptides (ANP and BNP) and beta-myosin heavy chain (β-MHC). Here we characterize the cardiac phenotype in the PA mouse model by histological and echocardiography studies and identify a series of upregulated cardiac-enriched microRNAs (miRNAs) in the PA mouse heart, some of them also altered as circulating miRNAs in PA patients' plasma samples. In PA mice hearts, we show alterations in signaling pathways regulated by the identified miRNAs, which could be contributing to cardiac remodeling and dysfunction; notably, an activation of the mammalian target of rapamycin (mTOR) pathway and a decrease in autophagy, which are reverted by rapamycin treatment. In vitro studies in HL-1 cardiomyocytes indicate that propionate, the major toxic metabolite accumulating in the disease, triggers the increase in expression levels of miRNAs, BNP, and β-MHC, concomitant with an increase in reactive oxygen species. Our results highlight miRNAs and signaling alterations in the PCC-deficient heart which may contribute to the development of PA-associated cardiomyopathy and provide a basis to identify new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Alejandro Fulgencio-Covián
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Esmeralda Alonso-Barroso
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | | | - Ana Rivera-Barahona
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Magdalena Ugarte
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain
| | - Belén Pérez
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | | | - Celia Pérez-Cerdá
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Eva Richard
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Lourdes R Desviat
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain.
| |
Collapse
|
85
|
Li XQ, Liu YK, Yi J, Dong JS, Zhang PP, Wan L, Li K. MicroRNA-143 Increases Oxidative Stress and Myocardial Cell Apoptosis in a Mouse Model of Doxorubicin-Induced Cardiac Toxicity. Med Sci Monit 2020; 26:e920394. [PMID: 32170053 PMCID: PMC7085239 DOI: 10.12659/msm.920394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Oxidative stress and myocardial apoptosis are features of doxorubicin-induced cardiac toxicity that can result in cardiac dysfunction. Previous studies showed that microRNA-143 (miR-143) was expressed in the myocardium and had a role in cardiac function. This study aimed to investigate the effects and possible molecular mechanisms of miR-143 on oxidative stress and myocardial cell apoptosis in a mouse model of doxorubicin-induced cardiac toxicity. MATERIAL AND METHODS Mice underwent intraperitoneal injection of doxorubicin (15 mg/kg) daily for eight days to develop the mouse model of doxorubicin-induced cardiac toxicity. Four days before doxorubicin administration, a group of mice was pretreated daily with a miR-143 antagonist (25 mg/kg/day) for four consecutive days by tail vein injection. The study included the use of a miR-143 antagomir, or anti-microRNA, an oligonucleotide that silenced endogenous microRNA (miR), and an agomir to miR-143, and also the AKT inhibitor, MK2206. Quantitative real-time polymerase chain reaction (qRT-PCR) and immunoblot analysis were used to measure mRNA and protein expression, respectively. RESULTS Doxorubicin treatment increased the expression of miR-143, which was reduced by the miR-143 antagomir. Overexpression of miR-143 increased doxorubicin-induced myocardial apoptosis and oxidative stress. The use of the miR-143 antagomir significantly activated protein kinase B (PKB) and AKT, which were reduced in the presence of the AKT inhibitor, MK2206. However, the use of the miR-143 antagomir further down-regulated AKT phosphorylation following doxorubicin treatment and increased AKT activation. CONCLUSIONS In a mouse model of doxorubicin-induced cardiac toxicity, miR-143 increased oxidative stress and myocardial cell apoptosis following doxorubicin treatment by inhibiting AKT.
Collapse
|
86
|
Resveratrol and Diabetic Cardiomyopathy: Focusing on the Protective Signaling Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7051845. [PMID: 32256959 PMCID: PMC7094200 DOI: 10.1155/2020/7051845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/01/2020] [Accepted: 02/17/2020] [Indexed: 02/07/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a common cardiovascular complication of diabetic mellitus that is characterized by diastolic disorder in the early stage and clinical heart failure in the later stage. Presently, DCM is considered one of the major causes of death in diabetic patients. Resveratrol (RSV), a naturally occurring stilbene, is widely reported as a cardioprotective substance in many heart diseases. Thus far, the specific roles of RSV in DCM prevention and treatment have attracted great attention. Here, we discuss the roles of RSV in DCM by focusing its downstream targets from both in vivo and in vitro studies. Among such targets, Sirtuins 1/3 and AMP-activated kinase have been identified as key mediators that induce cardioprotection during hyperglycemia. In addition, many other signaling molecules (e.g., forkhead box-O3a and extracellular regulated protein kinases) are also regulated in the presence of RSV and exert beneficial effects such as opposing oxidative stress, inflammation, and apoptosis in cardiomyocytes exposed to high-glucose conditions. The beneficial potential of an RSV/stem cell cotherapy is also reviewed as a promising therapeutic strategy for preventing the development of DCM.
Collapse
|
87
|
Ba RQ, Liu J, Fan XJ, Jin GL, Huang BG, Liu MW, Yang JS. Effects of miR-199a on autophagy by targeting glycogen synthase kinase 3β to activate PTEN/AKT/mTOR signaling in an MPP+ in vitro model of Parkinson’s disease. Neurol Res 2020; 42:308-318. [PMID: 32151238 DOI: 10.1080/01616412.2020.1726584] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Rui-Qiong Ba
- Department of Neurology, First People’s Hospital of Qujing City, Qujing City, China
| | - Jia Liu
- Department of Neurology, First People’s Hospital of Qujing City, Qujing City, China
| | - Xi-Jun Fan
- Department of Neurology, First People’s Hospital of Qujing City, Qujing City, China
| | - Gui-Li Jin
- Department of Neurology, First People’s Hospital of Qujing City, Qujing City, China
| | - Bao-Gang Huang
- Department of Neurology, First People’s Hospital of Qujing City, Qujing City, China
| | - Ming-Wei Liu
- Department of Emergency, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun-Su Yang
- Department of Neurology, First People’s Hospital of Qujing City, Qujing City, China
| |
Collapse
|
88
|
Gan X, Zhu H, Jiang X, Obiegbusi SC, Yong M, Long X, Hu J. CircMUC16 promotes autophagy of epithelial ovarian cancer via interaction with ATG13 and miR-199a. Mol Cancer 2020; 19:45. [PMID: 32111227 PMCID: PMC7047414 DOI: 10.1186/s12943-020-01163-z] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/13/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Circular RNA (circRNA) has been proven to play a significant role in multiple types of cancer. However, the expression and role of circRNAs in epithelial ovarian cancer (EOC) remains elusive. METHODS CircRNA and mRNA expression profiles of EOC were screened with sequencing analysis. Gene silencing and over-expression were used to study circRNA function. Cell proliferation and Matrigel invasion assays were used to detect cell proliferation and invasion, respectively. The expression of circRNAs, mRNAs and miRNAs was detected using qPCR. The location of circRNAs was detected using FISH. The expression of proteins was detected using western blot and immunohistochemistry. RESULTS CircMUC16 had increased expression in EOC tissues as compared to healthy ovarian tissues. The expression of circMUC16 was linked to the progression in stage and grade of EOC. Hence, silencing circMUC16 suppressed autophagy flux of SKOV3 cells. In contrast, ectopic expression of circMUC16 promoted autophagy flux of A2780 cells. CircMUC16-mediated autophagy exacerbated EOC invasion and metastasis. Mechanistically, circMUC16 could directly bind to miR-199a-5p and relieve suppression of target Beclin1 and RUNX1. In turn, RUNX1 elevated the expression of circMUC16 via promotion of its transcription. CircMUC16 could directly bind to ATG13 and promote its expression. CONCLUSION This study demonstrated that circMUC16 regulated Beclin1 and RUNX1 by sponging miR-199a-5p. The data suggested that circMUC16 could be a potential target for EOC diagnosis and therapy.
Collapse
Affiliation(s)
- Xiaoling Gan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hongtao Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xingwei Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Samuel C Obiegbusi
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Min Yong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xingtao Long
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, 400030, People's Republic of China
| | - Jianguo Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
89
|
Zhu H, Li Y, Wang MX, Wang JH, Du WX, Zhou F. Analysis of cardiovascular disease-related NF-κB-regulated genes and microRNAs in TNFα-treated primary mouse vascular endothelial cells. J Zhejiang Univ Sci B 2020; 20:803-815. [PMID: 31489800 DOI: 10.1631/jzus.b1800631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Activated nuclear factor-κB (NF-κB) plays an important role in the development of cardiovascular disease (CVD) through its regulated genes and microRNAs (miRNAs). However, the gene regulation profile remains unclear. In this study, primary mouse vascular endothelial cells (pMVECs) were employed to detect CVD-related NF-κB-regulated genes and miRNAs. Genechip assay identified 77 NF-κB-regulated genes, including 45 upregulated and 32 downregulated genes, in tumor necrosis factor α (TNFα)-treated pMVECs. Ten of these genes were also found to be regulated by NF-κB in TNFα-treated HeLa cells. Quantitative real-time PCR (qRT-PCR) assay confirmed the up-regulation of Egr1, Tnf, and Btg2 by NF-κB in the TNFα-treated pMVECs. The functional annotation revealed that many NF-κB-regulated genes identified in pMVECs were clustered into classical NF-κB-involved biological processes. Genechip assay also identified 26 NF-κB-regulated miRNAs, of which 21 were upregulated and 5 downregulated, in the TNFα-treated pMVECs. Further analysis showed that nine of the identified genes are regulated by seven of these miRNAs. Finally, among the identified NF-κB-regulated genes and miRNAs, 5 genes and 12 miRNAs were associated with CVD by miRWalk and genetic association database analysis. Taken together, these findings show an intricate gene regulation network raised by NF-κB in TNFα-treated pMVECs. The network provides new insights for understanding the molecular mechanism underlying the progression of CVD.
Collapse
Affiliation(s)
- Hui Zhu
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou 521041, China
| | - Yun Li
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou 521041, China
| | - Mao-Xian Wang
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou 521041, China
| | - Ju-Hong Wang
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou 521041, China
| | - Wen-Xin Du
- Shandong Center for Drug and Food Evaluation & Certification, Jinan 250014, China
| | - Fei Zhou
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou 521041, China
| |
Collapse
|
90
|
Chen Y, Hua Y, Li X, Arslan IM, Zhang W, Meng G. Distinct Types of Cell Death and the Implication in Diabetic Cardiomyopathy. Front Pharmacol 2020; 11:42. [PMID: 32116717 PMCID: PMC7018666 DOI: 10.3389/fphar.2020.00042] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 01/14/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a chronic complication of diabetes mellitus, characterized by abnormalities of myocardial structure and function. Researches on the models of type 1 and type 2 diabetes mellitus as well as the application of genetic engineering technology help in understanding the molecular mechanism of DCM. DCM has multiple hallmarks, including hyperglycemia, insulin resistance, increased free radical production, lipid peroxidation, mitochondrial dysfunction, endothelial dysfunction, and cell death. Essentially, cell death is considered to be the terminal pathway of cardiomyocytes during DCM. Morphologically, cell death can be classified into four different forms: apoptosis, autophagy, necrosis, and entosis. Apoptosis, as type I cell death, is the fastest form of cell death and mainly occurs depending on the caspase proteolytic cascade. Autophagy, as type II cell death, is a degradation process to remove damaged proteins, dysfunctional organelles and commences by the formation of autophagosome. Necrosis is type III cell death, which contains a great diversity of cell death processes, such as necroptosis and pyroptosis. Entosis is type IV cell death, displaying “cell-in-cell” cytological features and requires the engulfing cells to execute. There are also some other types of cell death such as ferroptosis, parthanatos, netotic cell death, lysosomal dependent cell death, alkaliptosis or oxeiptosis, which are possibly involved in DCM. Drugs or compounds targeting the signals involved in cell death have been used in clinics or experiments to treat DCM. This review briefly summarizes the mechanisms and implications of cell death in DCM, which is beneficial to improve the understanding of cell death in DCM and may propose novel and ideal strategies in future.
Collapse
Affiliation(s)
- Yun Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, China.,School of Medicine, Nantong University, Nantong, China
| | - Yuyun Hua
- Department of Pharmacology, School of Pharmacy, Nantong University, Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, China
| | - Xinshuai Li
- Department of Pharmacology, School of Pharmacy, Nantong University, Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, China
| | | | - Wei Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, China
| | - Guoliang Meng
- Department of Pharmacology, School of Pharmacy, Nantong University, Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, China.,School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
91
|
Kura B, Szeiffova Bacova B, Kalocayova B, Sykora M, Slezak J. Oxidative Stress-Responsive MicroRNAs in Heart Injury. Int J Mol Sci 2020; 21:E358. [PMID: 31948131 PMCID: PMC6981696 DOI: 10.3390/ijms21010358] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are important molecules in the living organisms as a part of many signaling pathways. However, if overproduced, they also play a significant role in the development of cardiovascular diseases, such as arrhythmia, cardiomyopathy, ischemia/reperfusion injury (e.g., myocardial infarction and heart transplantation), and heart failure. As a result of oxidative stress action, apoptosis, hypertrophy, and fibrosis may occur. MicroRNAs (miRNAs) represent important endogenous nucleotides that regulate many biological processes, including those involved in heart damage caused by oxidative stress. Oxidative stress can alter the expression level of many miRNAs. These changes in miRNA expression occur mainly via modulation of nuclear factor erythroid 2-related factor 2 (Nrf2), sirtuins, calcineurin/nuclear factor of activated T cell (NFAT), or nuclear factor kappa B (NF-κB) pathways. Up until now, several circulating miRNAs have been reported to be potential biomarkers of ROS-related cardiac diseases, including myocardial infarction, hypertrophy, ischemia/reperfusion, and heart failure, such as miRNA-499, miRNA-199, miRNA-21, miRNA-144, miRNA-208a, miRNA-34a, etc. On the other hand, a lot of studies are aimed at using miRNAs for therapeutic purposes. This review points to the need for studying the role of redox-sensitive miRNAs, to identify more effective biomarkers and develop better therapeutic targets for oxidative-stress-related heart diseases.
Collapse
Affiliation(s)
- Branislav Kura
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Barbara Szeiffova Bacova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Barbora Kalocayova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Matus Sykora
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Jan Slezak
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| |
Collapse
|
92
|
|
93
|
Jin L, Zhou Y, Han L, Piao J. MicroRNA302-367-PI3K-PTEN-AKT-mTORC1 pathway promotes the development of cardiac hypertrophy through controlling autophagy. In Vitro Cell Dev Biol Anim 2019; 56:112-119. [PMID: 31845077 DOI: 10.1007/s11626-019-00417-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022]
Abstract
Cardiac hypertrophy at a decompensated state eventually leads to heart failure that mostly contributes to deaths globally. Dysregulated cardiac autophagy is a hallmark of a diseased heart, and a close contact between cardiac autophagy and cardiac hypertrophy is emerging. MicroRNAs (miRNAs) have been recently reported to be prominently implicated in cardiac hypertrophy through regulating cardiac autophagy. However, the role and function of miR302-367 clusters in cardiac autophagy and cardiac hypertrophy remain largely masked. Therefore, to investigate the performance of miR302-367 in cardiac hypertrophy, the specific in vitro hypertrophic model was established in H9c2 cells upon Ang II treatment. Consequently, we discovered a distinct inhibition on autophagy and a remarkable upregulation of miR302-367 expression in hypertrophic H9c2 cells. Besides, loss- and gain-of-function assays demonstrated miR302-367 inhibited autophagy and then aggravated cardiac hypertrophy. Mechanically, PTEN was predicted and confirmed as the shared target of miR302-367. Further, we recognized the apparent inactivation of PI3K/AKT/mTORC1 signaling in the face of miR302-367 suppression in Ang II-induced hypertrophic H9c2 cells. Moreover, co-treatment of PTEN inhibitor re-activated the PI3K/AKT/mTORC1 pathway, therefore counteracting the pro-autophagic and anti-hypertrophic effects of miR302-367 depletion on cardiomyocytes. These findings unveiled the pivotal role of the miR302-367 cluster in regulating cardiac autophagy and therefore modulating cardiac hypertrophy through PTEN/PI3K/AKT/mTORC1 signaling, indicating a promising therapeutic strategy for cardiac hypertrophy and even heart failure. Graphical abstract .
Collapse
Affiliation(s)
- Lianhua Jin
- Cardiology Department of Pediatric of the First Hospital of Jilin University, No.71 Xinmin Street, Changchun City, 130021, Jilin Province, China
| | - Yan Zhou
- Cardiology Department of Pediatric of the First Hospital of Jilin University, No.71 Xinmin Street, Changchun City, 130021, Jilin Province, China
| | - Lizhi Han
- Cardiology Department of Pediatric of the First Hospital of Jilin University, No.71 Xinmin Street, Changchun City, 130021, Jilin Province, China
| | - Jinhua Piao
- Cardiology Department of Pediatric of the First Hospital of Jilin University, No.71 Xinmin Street, Changchun City, 130021, Jilin Province, China.
| |
Collapse
|
94
|
Wang Q, Ye B, Wang P, Yao F, Zhang C, Yu G. Overview of microRNA-199a Regulation in Cancer. Cancer Manag Res 2019; 11:10327-10335. [PMID: 31849522 PMCID: PMC6911337 DOI: 10.2147/cmar.s231971] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/23/2019] [Indexed: 12/17/2022] Open
Abstract
microRNAs (miRNAs) are a class of endogenous short, non-coding RNAs that regulate a multitude of genes at the post-transcriptional level. miR-199, which is a highly conserved miRNA family, consists of miR-199a and miR-199b. Researchers mainly focused on miR-199a over the past few years. Functional studies have demonstrated that mature miR-199a is a key player in the maintenance of normal homeostasis and in the regulation of disease pathogenesis. Here, we summarize the biological functions of miR-199a and review recent research on its roles in the physiological processes of cancer cells, such as proliferation, migration, invasion, apoptosis, autophagy and glycometabolism.
Collapse
Affiliation(s)
- Qiwen Wang
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang 453007, Henan, People's Republic of China.,State Key Laboratory Cell Differentiation and Regulation, College of Life Science, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Bingyu Ye
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang 453007, Henan, People's Republic of China.,State Key Laboratory Cell Differentiation and Regulation, College of Life Science, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Ping Wang
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang 453007, Henan, People's Republic of China.,State Key Laboratory Cell Differentiation and Regulation, College of Life Science, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Fenjie Yao
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang 453007, Henan, People's Republic of China.,State Key Laboratory Cell Differentiation and Regulation, College of Life Science, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Chunyan Zhang
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang 453007, Henan, People's Republic of China.,State Key Laboratory Cell Differentiation and Regulation, College of Life Science, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Guoying Yu
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang 453007, Henan, People's Republic of China.,State Key Laboratory Cell Differentiation and Regulation, College of Life Science, Henan Normal University, Xinxiang 453007, People's Republic of China
| |
Collapse
|
95
|
Qiang Z, Jin B, Peng Y, Zhang Y, Wang J, Chen C, Wang X, Liu F. miR-762 modulates thyroxine-induced cardiomyocyte hypertrophy by inhibiting Beclin-1. Endocrine 2019; 66:585-595. [PMID: 31522342 DOI: 10.1007/s12020-019-02048-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/05/2019] [Indexed: 01/07/2023]
Abstract
PURPOSE Whether autophagy plays a key role in thyroxine-induced cardiomyocyte hypertrophy, and whether the role of autophagy in thyroxine-induced cardiomyocyte hypertrophy is related to targeting of Beclin-1 by miR-762 remains unclear. This research focused on testing these two hypotheses. Importantly, the results of this study will help us better understand the molecular mechanisms of thyroxine-induced cardiomyocyte hypertrophy. METHODS In vivo and in vitro, RT-PCR, western blot, and dual luciferase reporter assay were performed to understand the molecular mechanism of thyroxine-induced cardiomyocyte hypertrophy. HE staining, Masson staining, transmission electron microscopy, and immunofluorescence were used to observe intuitively changes of hearts and cardiomyocytes. RESULTS Our results showed that in vivo, serum TT3, TT4, and heart rate were significantly upregulated in the T4 group compared with the control group. Moreover, the surface area of cardiomyocytes was significantly increased in the T4 group, and the structural disorder was accompanied by obvious hyperplasia of collagen fibers. The expression of ANP, and β-MHC was significantly upregulated in the T4 group. In addition, LC3 II/LC3 I, Beclin-1 and the count of autophagic vacuoles were significantly upregulated, but miR-762 was significantly downregulated in the T4 group compared to the control group. Subsequently, a dual luciferase reporter assay suggested that Beclin-1 was the target gene of miR-762. In vitro, the results for the T3 group were consistent with the results for the T4 group. Furthermore, cardiomyocyte hypertrophy and autophagic activity were attenuated in the T3 + miR-762 mimic group compared with the T3 group. In contrast, cardiomyocyte hypertrophy and autophagic activity were aggravated in the T3 + miR-762 inhibitor group compared with the T3 group. CONCLUSIONS miR-762 modulates thyroxine-induced cardiomyocyte hypertrophy by inhibiting Beclin-1.
Collapse
Affiliation(s)
- Zheng Qiang
- Department of Anatomy, Guilin Medical University, 541004, Guilin, China.
| | - Beifang Jin
- Department of Anatomy, Guilin Medical University, 541004, Guilin, China
| | - Yuntao Peng
- Department of Anatomy, Guilin Medical University, 541004, Guilin, China
| | - Yan Zhang
- Department of Anatomy, Guilin Medical University, 541004, Guilin, China
| | - Junfeng Wang
- Department of Anatomy, Guilin Medical University, 541004, Guilin, China
| | - Chen Chen
- Department of Anatomy, Guilin Medical University, 541004, Guilin, China
| | - Xinfeng Wang
- Department of Anatomy, Guilin Medical University, 541004, Guilin, China
| | - Fang Liu
- Department of Anatomy, Guilin Medical University, 541004, Guilin, China.
- Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, 541004, Guilin, China.
| |
Collapse
|
96
|
Bonora M, Wieckowski MR, Sinclair DA, Kroemer G, Pinton P, Galluzzi L. Targeting mitochondria for cardiovascular disorders: therapeutic potential and obstacles. Nat Rev Cardiol 2019; 16:33-55. [PMID: 30177752 DOI: 10.1038/s41569-018-0074-0] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A large body of evidence indicates that mitochondrial dysfunction has a major role in the pathogenesis of multiple cardiovascular disorders. Over the past 2 decades, extraordinary efforts have been focused on the development of agents that specifically target mitochondria for the treatment of cardiovascular disease. Despite such an intensive wave of investigation, no drugs specifically conceived to modulate mitochondrial functions are currently available for the clinical management of cardiovascular disease. In this Review, we discuss the therapeutic potential of targeting mitochondria in patients with cardiovascular disease, examine the obstacles that have restrained the development of mitochondria-targeting agents thus far, and identify strategies that might empower the full clinical potential of this approach.
Collapse
Affiliation(s)
- Massimo Bonora
- Ruth L. and David S. Gottesman Institute for Stem Cell, Regenerative Medicine Research, Department of Cell Biology and Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mariusz R Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - David A Sinclair
- Department of Genetics, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA.,Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Guido Kroemer
- Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Paolo Pinton
- Department of Morphology, Surgery, and Experimental Medicine, Section of Pathology, Oncology, and Experimental Biology, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy. .,Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, Italy.
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Paris, France. .,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA. .,Sandra and Edward Meyer Cancer Center, New York, NY, USA.
| |
Collapse
|
97
|
Wang S, Sun H, Zhan X, Wang Q. MicroRNA‑718 serves a tumor‑suppressive role in non‑small cell lung cancer by directly targeting CCNB1. Int J Mol Med 2019; 45:33-44. [PMID: 31746372 PMCID: PMC6889928 DOI: 10.3892/ijmm.2019.4396] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 08/13/2019] [Indexed: 12/19/2022] Open
Abstract
MicroRNA‑718 (miR‑718) serves crucial roles in tumorigenesis and in the progression of a number of cancers. However, the expression profile, specific functions and mechanisms of action of miR‑718 in non‑small cell lung cancer (NSCLC) are still elusive. The aims of the present study were to quantify the expression of miR‑718, determine its biological roles and elucidate the molecular mechanisms responsible for its activities in NSCLC cells. Reverse transcription‑quantitative PCR was carried out to assess miR‑718 expression in NSCLC tissue samples and cell lines. The Cell Counting Kit‑8 assay, flow cytometry, cell migration and invasion assays, and a tumor xenograft experiment were performed to evaluate the effects of miR‑718 overexpression on the malignant biological behaviors of NSCLC cells. miR‑718 expression was demonstrated to be significantly decreased in NSCLC tissue samples and cell lines. This reduced expression was significantly associated with tumor, node, metastasis stage, tumor size, lymph node metastasis and poor overall survival among patients with NSCLC. Exogenous miR‑718 expression suppressed NSCLC cell proliferation, migration and invasion, and promoted apoptosis in vitro; whereas it hindered tumor growth in vivo. Experiments to elucidate the mechanisms involved revealed that miR‑718 functions by directly targeting cyclin B1 (CCNB1) mRNA. CCNB1 expression was found to be upregulated in NSCLC and inversely correlated with miR‑718 levels. CCNB1 depletion had effects similar to those of miR‑718 overexpression in NSCLC cells. Furthermore, restoration of CCNB1 expression attenuated the tumor‑suppressive effects of miR‑718 overexpression in NSCLC cells. These results indicated that miR‑718 suppressed NSCLC progression in vitro and in vivo by directly targeting CCNB1 mRNA, which may indicate a potential target for the diagnosis and treatment of this fatal disease.
Collapse
Affiliation(s)
- Shu Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Hongmei Sun
- Department of Thoracic Oncosurgery, Jilin Province Tumor Hospital, Changchun, Jilin 130012, P.R. China
| | - Xiaokai Zhan
- Department of Thoracic Oncosurgery, Jilin Province Tumor Hospital, Changchun, Jilin 130012, P.R. China
| | - Qiwen Wang
- Department of Thoracic Oncosurgery, Jilin Province Tumor Hospital, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
98
|
Zhao Y, Wang Z, Zhang W, Zhang L. MicroRNAs play an essential role in autophagy regulation in various disease phenotypes. Biofactors 2019; 45:844-856. [PMID: 31418958 PMCID: PMC6916288 DOI: 10.1002/biof.1555] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 07/31/2019] [Indexed: 12/16/2022]
Abstract
Autophagy is a highly conserved catabolic process and fundamental biological process in eukaryotic cells. It recycles intracellular components to provide nutrients during starvation and maintains quality control of organelles and proteins. In addition, autophagy is a well-organized homeostatic cellular process that is responsible for the removal of damaged organelles and intracellular pathogens. Moreover, it also modulates the innate and adaptive immune systems. Micro ribonucleic acids (microRNAs) are a mature class of post-transcriptional modulators that are widely expressed in tissues and organs. And, it can suppress gene expression by targeting messenger RNAs for translational repression or, at a lesser extent, degradation. Research indicates that microRNAs regulate autophagy through different pathways, playing an essential role in the treatment of various diseases. It is an important regulator of fundamental cellular processes such as proliferation, autophagy, and cell apoptosis. In this review article, we first review the current knowledge of autophagy and the function of microRNAs. Then, we summarize the mechanism of autophagy and the signaling pathways related to autophagy by citing at least the main proteins involved in the different phases of the process. Second, we introduce other members of RNA and report some examples in various pathologies. Finally, we review the current literature regarding microRNA-based therapies for cancer, atherosclerosis, cardiac disease, tuberculosis, and viral diseases. MicroRNAs can cause autophagy upregulation or downregulation by targeting genes or affecting autophagy-related signaling pathways. Therefore, the microRNAs have a huge potential in autophagy regulation, and it is the function as diagnostic and prognostic markers.
Collapse
Affiliation(s)
- Yunyi Zhao
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
| | - Ze Wang
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
| | - Wenhui Zhang
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
- Ministry of Education, Engineering Research Center for Bioreactor and Pharmaceutical DevelopmentJilin Agricultural UniversityChangchunChina
| | - Linbo Zhang
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
- Ministry of Education, Engineering Research Center for Bioreactor and Pharmaceutical DevelopmentJilin Agricultural UniversityChangchunChina
| |
Collapse
|
99
|
Abstract
Cardiovascular diseases are the most prominent maladies in aging societies. Indeed, aging promotes the structural and functional declines of both the heart and the blood circulation system. In this review, we revise the contribution of known longevity pathways to cardiovascular health and delineate the possibilities to interfere with them. In particular, we evaluate autophagy, the intracellular catabolic recycling system associated with life- and health-span extension. We present genetic models, pharmacological interventions, and dietary strategies that block, reduce, or enhance autophagy upon age-related cardiovascular deterioration. Caloric restriction or caloric restriction mimetics like metformin, spermidine, and rapamycin (all of which trigger autophagy) are among the most promising cardioprotective interventions during aging. We conclude that autophagy is a fundamental process to ensure cardiac and vascular health during aging and outline its putative therapeutic importance.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- From the Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.)
| | - Simon Sedej
- From the Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.).,BioTechMed Graz, Austria (S.S., D.C.-G., F.M.)
| | - Didac Carmona-Gutierrez
- BioTechMed Graz, Austria (S.S., D.C.-G., F.M.).,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria (D.C.-G., F.M.)
| | - Frank Madeo
- BioTechMed Graz, Austria (S.S., D.C.-G., F.M.).,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria (D.C.-G., F.M.)
| | - Guido Kroemer
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France (G.K.).,Cell Biology and Metabolomics Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France (G.K.).,INSERM, U1138, Paris, France (G.K.).,Université Paris Descartes, Sorbonne Paris Cité, France (G.K.).,Université Pierre et Marie Curie, Paris, France (G.K.).,Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France (G.K.).,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden (G.K.)
| |
Collapse
|
100
|
Cao Y, Wen J, Li Y, Chen W, Wu Y, Li J, Huang G. Uric acid and sphingomyelin enhance autophagy in iPS cell-originated cardiomyocytes through lncRNA MEG3/miR-7-5p/EGFR axis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3774-3785. [PMID: 31559872 DOI: 10.1080/21691401.2019.1667817] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yinyin Cao
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai, PR China
| | - Junxiang Wen
- Clinical Laboratory Center, Children’s Hospital of Fudan University, Shanghai, PR China
| | - Yang Li
- Clinical Laboratory Center, Children’s Hospital of Fudan University, Shanghai, PR China
| | - Weicheng Chen
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai, PR China
| | - Yao Wu
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai, PR China
| | - Jian Li
- Clinical Laboratory Center, Children’s Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, PR China
| | - Guoying Huang
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, PR China
| |
Collapse
|