1
|
Sopić M, Vladimirov S, Munjas J, Mitić T, Hall IF, Jusic A, Ruzic D, Devaux Y. Targeting noncoding RNAs to treat atherosclerosis. Br J Pharmacol 2025; 182:220-245. [PMID: 38720437 DOI: 10.1111/bph.16412] [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: 10/26/2023] [Revised: 03/06/2024] [Accepted: 04/05/2024] [Indexed: 12/13/2024] Open
Abstract
Noncoding RNAs (ncRNAs) are pivotal for various pathological processes, impacting disease progression. The potential for leveraging ncRNAs to prevent or treat atherosclerosis and associated cardiovascular diseases is of great significance, especially given the increasing prevalence of atherosclerosis in an ageing and sedentary population. Together, these diseases impose a substantial socio-economic burden, demanding innovative therapeutic solutions. This review explores the potential of ncRNAs in atherosclerosis treatment. We commence by examining approaches for identifying and characterizing atherosclerosis-associated ncRNAs. We then delve into the functional aspects of ncRNAs in atherosclerosis development and progression. Additionally, we review current RNA and RNA-targeting molecules in development or under approval for clinical use, offering insights into their pharmacological potential. The importance of improved ncRNA delivery strategies is highlighted. Finally, we suggest avenues for advanced research to accelerate the use of ncRNAs in treating atherosclerosis and mitigating its societal impact. LINKED ARTICLES: This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
Affiliation(s)
- Miron Sopić
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Sandra Vladimirov
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Jelena Munjas
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Tijana Mitić
- BHF/University Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ignacio Fernando Hall
- BHF/University Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Amela Jusic
- HAYA Therapeutics SA, SuperLab Suisse - Bâtiment Serine, Lausanne, Vaud, Switzerland
| | - Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
Collapse
|
2
|
Honda T, Sakai H, Inui M. Intracellular delivery of a phospholamban-targeting aptamer using cardiomyocyte-internalizing aptamers. Eur J Pharmacol 2024; 985:177130. [PMID: 39536855 DOI: 10.1016/j.ejphar.2024.177130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
The sarco (endo)plasmic reticulum Ca2+-ATPase 2a (SERCA2a)-phospholamban (PLN) system within the sarcoplasmic reticulum is crucial for regulating intracellular Ca2+ cycling in ventricular cardiomyocytes. Given that impaired Ca2+ cycling is associated with heart failure, modulating SERCA2a activity represents a promising therapeutic strategy. Previously, we engineered an RNA aptamer (Apt30) that binds to PLN, thereby activating SERCA2a by alleviating PLN's inhibitory effect. However, Apt30 alone cannot reach intracellular PLN, necessitating the development of a mechanism for its specific internalization into cardiomyocytes. Using the systematic evolution of ligands by exponential enrichment (SELEX) method, we isolated RNA aptamers capable of internalizing into cardiomyocytes. These aptamers demonstrated sub-micromolar EC50 values for cardiomyocyte internalization and exhibited significantly reduced activity against various non-myocardial cells, highlighting their specificity for cardiomyocytes. Moreover, some of these cardiomyocyte-internalizing aptamers could be linked to Apt30 as a single RNA strand without compromising their internalization efficacy. Supplementing the culture medium with these hybrid aptamers enhanced Ca2+ transients and contractile function in rat cardiomyocytes. These findings provide critical insights for developing novel therapeutics directly acting on PLN in cardiomyocytes, potentially compensating for the disadvantages of conventional methods that involve viral vector-mediated intracellular transduction or alterations in endogenous protein expression.
Collapse
Affiliation(s)
- Takeshi Honda
- Department of Pharmacology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan.
| | - Hiroki Sakai
- Department of Pharmacology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Makoto Inui
- Department of Pharmacology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| |
Collapse
|
3
|
Saddique MN, Qadri M, Ain NU, Farhan E, Shahid F, Benyamin J, Bashir MA, Jain H, Iqbal J. Safety and effectiveness of interference RNA (RNAi) based therapeutics in cardiac failure: A systematic review. Heart Lung 2024; 68:298-304. [PMID: 39214039 DOI: 10.1016/j.hrtlng.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Heart failure is a major worldwide health concern and leading cause of mortality. RNAi interventions hold promise for patients resistant to conventional drugs due to their off-target effects and lack of specificity. OBJECTIVES To examine the safety and effectiveness of RNAi therapeutics in treating heart failure. METHODS The PubMed, Embase, Scopus and Cochrane databases were searched using appropriate keyword from inception until December 31, 2023. A total of 14 studies fulfilling predefined selection criteria were included for qualitative synthesis. RESULTS We found that in patients with cardiac amyloidosis, patisiran and revusiran showed considerable improvements in cardiac output and left ventricular wall thickness. In animal studies, Nox2-siRNA showed effectiveness in regaining heart function. Furthermore, cardiomyocyte count and left ventricular function were improved by DUSP5 siRNA + T3 therapy and meg3 inhibition after myocardial infarction (MI). RNAi showed minimal adverse effects like peripheral neuropathy, hepatotoxicity, urinary tract infection, vaginal infection, diarrhea, abdominal pain arrhythmias, conduction disorders, and cardiotoxicity (LV wall thinning, heart failure) and improved cardiac biomarkers. CONCLUSION RNAi therapeutics are novel treatment option for improving cardiac function because their high target specificity, ability to target genes that conventional drugs struggle to reach and potential for long-lasting effects. Further research on optimizing delivery methods, improving target specificity, evaluating long-term safety profiles and cost-effectiveness to fully realize their potential.
Collapse
Affiliation(s)
| | - Maria Qadri
- Jinnah Sindh Medical University, Karachi 75510, Pakistan
| | - Noor Ul Ain
- FMH College of Medicine and Dentistry, Lahore 54000, Pakistan
| | - Eesha Farhan
- Karachi Institute of Medical Sciences, Karachi 75510, Pakistan
| | - Fatima Shahid
- King Edward Medical University, Lahore 54000, Pakistan
| | | | | | - Hritvik Jain
- Department of Internal Medicine, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Javed Iqbal
- Nursing Department, Communicable Disease Center-Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
4
|
Vora N, Patel P, Gajjar A, Ladani P, Konat A, Bhanderi D, Gadam S, Prajjwal P, Sharma K, Arunachalam SP. Gene therapy for heart failure: A novel treatment for the age old disease. Dis Mon 2024; 70:101636. [PMID: 37734966 DOI: 10.1016/j.disamonth.2023.101636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Across the globe, cardiovascular disease (CVD) is the leading cause of mortality. According to reports, around 6.2 million people in the United states have heart failure. Current standards of care for heart failure can delay but not prevent progression of disease. Gene therapy is one of the novel treatment modalities that promises to fill this limitation in the current standard of care for Heart Failure. In this paper we performed an extensive search of the literature on various advances made in gene therapy for heart failure till date. We review the delivery methods, targets, current applications, trials, limitations and feasibility of gene therapy for heart failure. Various methods have been employed till date for administering gene therapies including but not limited to arterial and venous infusion, direct myocardial injection and pericardial injection. Various strategies such as AC6 expression, S100A1 protein upregulation, VEGF-B and SDF-1 gene therapy have shown promise in recent preclinical trials. Furthermore, few studies even show that stimulation of cardiomyocyte proliferation such as through cyclin A2 overexpression is a realistic avenue. However, a considerable number of obstacles need to be overcome for gene therapy to be part of standard treatment of care such as definitive choice of gene, gene delivery systems and a suitable method for preclinical trials and clinical trials on patients. Considering the challenges and taking into account the recent advances in gene therapy research, there are encouraging signs to indicate gene therapy for heart failure to be a promising treatment modality for the future. However, the time and feasibility of this option remains in a situation of balance.
Collapse
Affiliation(s)
- Neel Vora
- B. J. Medical College, Ahmedabad, India
| | - Parth Patel
- Pramukhswami Medical College, Karamsad, India
| | | | | | - Ashwati Konat
- University School of Sciences, Gujarat University, Ahmedabad, India
| | | | | | | | - Kamal Sharma
- U. N. Mehta Institute of Cardiology and Research Centre, Ahmedabad, India.
| | | |
Collapse
|
5
|
Mattiazzi A, Kranias EG. Unleashing the Power of Genetics: PLN Ablation, Phospholambanopathies and Evolving Challenges. Circ Res 2024; 134:138-142. [PMID: 38236951 DOI: 10.1161/circresaha.123.323053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Affiliation(s)
- Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares, Centro Cientifico Tecnologico-La Plata CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Argentina (A.M.)
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, OH (E.G.K.)
| |
Collapse
|
6
|
Poller W, Sahoo S, Hajjar R, Landmesser U, Krichevsky AM. Exploration of the Noncoding Genome for Human-Specific Therapeutic Targets-Recent Insights at Molecular and Cellular Level. Cells 2023; 12:2660. [PMID: 37998395 PMCID: PMC10670380 DOI: 10.3390/cells12222660] [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: 10/06/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
While it is well known that 98-99% of the human genome does not encode proteins, but are nevertheless transcriptionally active and give rise to a broad spectrum of noncoding RNAs [ncRNAs] with complex regulatory and structural functions, specific functions have so far been assigned to only a tiny fraction of all known transcripts. On the other hand, the striking observation of an overwhelmingly growing fraction of ncRNAs, in contrast to an only modest increase in the number of protein-coding genes, during evolution from simple organisms to humans, strongly suggests critical but so far essentially unexplored roles of the noncoding genome for human health and disease pathogenesis. Research into the vast realm of the noncoding genome during the past decades thus lead to a profoundly enhanced appreciation of the multi-level complexity of the human genome. Here, we address a few of the many huge remaining knowledge gaps and consider some newly emerging questions and concepts of research. We attempt to provide an up-to-date assessment of recent insights obtained by molecular and cell biological methods, and by the application of systems biology approaches. Specifically, we discuss current data regarding two topics of high current interest: (1) By which mechanisms could evolutionary recent ncRNAs with critical regulatory functions in a broad spectrum of cell types (neural, immune, cardiovascular) constitute novel therapeutic targets in human diseases? (2) Since noncoding genome evolution is causally linked to brain evolution, and given the profound interactions between brain and immune system, could human-specific brain-expressed ncRNAs play a direct or indirect (immune-mediated) role in human diseases? Synergistic with remarkable recent progress regarding delivery, efficacy, and safety of nucleic acid-based therapies, the ongoing large-scale exploration of the noncoding genome for human-specific therapeutic targets is encouraging to proceed with the development and clinical evaluation of novel therapeutic pathways suggested by these research fields.
Collapse
Affiliation(s)
- Wolfgang Poller
- Department for Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum Charité (DHZC), Charité-Universitätsmedizin Berlin, 12200 Berlin, Germany;
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, 10785 Berlin, Germany
| | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA;
| | - Roger Hajjar
- Gene & Cell Therapy Institute, Mass General Brigham, 65 Landsdowne St, Suite 143, Cambridge, MA 02139, USA;
| | - Ulf Landmesser
- Department for Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum Charité (DHZC), Charité-Universitätsmedizin Berlin, 12200 Berlin, Germany;
- German Center for Cardiovascular Research (DZHK), Site Berlin, 10785 Berlin, Germany
- Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Anna M. Krichevsky
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
7
|
Aderinto N, Abdulbasit MO, Olatunji G, Edun M, Aboderin G. The promise of RNA-based therapeutics in revolutionizing heart failure management - a narrative review of current evidence. Ann Med Surg (Lond) 2023; 85:4442-4453. [PMID: 37663746 PMCID: PMC10473317 DOI: 10.1097/ms9.0000000000001118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 07/18/2023] [Indexed: 09/05/2023] Open
Abstract
This review elucidates the potential of RNA-based therapeutics to revolutionize heart failure (HF) management. Through a comprehensive analysis of relevant studies, this review reveals the promising prospects of these novel interventions in personalized treatment strategies, targeted modulation of specific molecular pathways, and the attainment of synergistic effects via combination therapies. Moreover, the regenerative capacity of RNA-based therapeutics for cardiac repair and the inherent advantages associated with noninvasive routes of administration are explored. Additionally, the studies accentuate the significance of diligent monitoring of disease progression and treatment response, ensuring safety and considering long-term outcomes. While ongoing research endeavours and technological advancements persist in addressing extant challenges and limitations, the transformative potential of RNA-based therapeutics in HF management offers a beacon of hope for enhanced patient outcomes.
Collapse
Affiliation(s)
- Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Oyo State
| | - Muili O. Abdulbasit
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Oyo State
| | - Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Mariam Edun
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Gbolahan Aboderin
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Oyo State
| |
Collapse
|
8
|
Innate Immunity in Cardiovascular Diseases-Identification of Novel Molecular Players and Targets. J Clin Med 2023; 12:jcm12010335. [PMID: 36615135 PMCID: PMC9821340 DOI: 10.3390/jcm12010335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
During the past few years, unexpected developments have driven studies in the field of clinical immunology. One driver of immense impact was the outbreak of a pandemic caused by the novel virus SARS-CoV-2. Excellent recent reviews address diverse aspects of immunological re-search into cardiovascular diseases. Here, we specifically focus on selected studies taking advantage of advanced state-of-the-art molecular genetic methods ranging from genome-wide epi/transcriptome mapping and variant scanning to optogenetics and chemogenetics. First, we discuss the emerging clinical relevance of advanced diagnostics for cardiovascular diseases, including those associated with COVID-19-with a focus on the role of inflammation in cardiomyopathies and arrhythmias. Second, we consider newly identified immunological interactions at organ and system levels which affect cardiovascular pathogenesis. Thus, studies into immune influences arising from the intestinal system are moving towards therapeutic exploitation. Further, powerful new research tools have enabled novel insight into brain-immune system interactions at unprecedented resolution. This latter line of investigation emphasizes the strength of influence of emotional stress-acting through defined brain regions-upon viral and cardiovascular disorders. Several challenges need to be overcome before the full impact of these far-reaching new findings will hit the clinical arena.
Collapse
|
9
|
Samra M, Srivastava K. Non-coding RNA and their potential role in cardiovascular diseases. Gene 2023; 851:147011. [DOI: 10.1016/j.gene.2022.147011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/27/2022] [Accepted: 10/21/2022] [Indexed: 11/27/2022]
|
10
|
Gast M, Nageswaran V, Kuss AW, Tzvetkova A, Wang X, Mochmann LH, Rad PR, Weiss S, Simm S, Zeller T, Voelzke H, Hoffmann W, Völker U, Felix SB, Dörr M, Beling A, Skurk C, Leistner DM, Rauch BH, Hirose T, Heidecker B, Klingel K, Nakagawa S, Poller WC, Swirski FK, Haghikia A, Poller W. tRNA-like Transcripts from the NEAT1-MALAT1 Genomic Region Critically Influence Human Innate Immunity and Macrophage Functions. Cells 2022; 11:cells11243970. [PMID: 36552736 PMCID: PMC9777231 DOI: 10.3390/cells11243970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/23/2022] [Accepted: 11/26/2022] [Indexed: 12/13/2022] Open
Abstract
The evolutionary conserved NEAT1-MALAT1 gene cluster generates large noncoding transcripts remaining nuclear, while tRNA-like transcripts (mascRNA, menRNA) enzymatically generated from these precursors translocate to the cytosol. Whereas functions have been assigned to the nuclear transcripts, data on biological functions of the small cytosolic transcripts are sparse. We previously found NEAT1-/- and MALAT1-/- mice to display massive atherosclerosis and vascular inflammation. Here, employing selective targeted disruption of menRNA or mascRNA, we investigate the tRNA-like molecules as critical components of innate immunity. CRISPR-generated human ΔmascRNA and ΔmenRNA monocytes/macrophages display defective innate immune sensing, loss of cytokine control, imbalance of growth/angiogenic factor expression impacting upon angiogenesis, and altered cell-cell interaction systems. Antiviral response, foam cell formation/oxLDL uptake, and M1/M2 polarization are defective in ΔmascRNA/ΔmenRNA macrophages, defining first biological functions of menRNA and describing new functions of mascRNA. menRNA and mascRNA represent novel components of innate immunity arising from the noncoding genome. They appear as prototypes of a new class of noncoding RNAs distinct from others (miRNAs, siRNAs) by biosynthetic pathway and intracellular kinetics. Their NEAT1-MALAT1 region of origin appears as archetype of a functionally highly integrated RNA processing system.
Collapse
Affiliation(s)
- Martina Gast
- Department of Cardiology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 12200 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, 12200 Berlin, Germany
| | - Vanasa Nageswaran
- Department of Cardiology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 12200 Berlin, Germany
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 12200 Berlin, Germany
| | - Andreas W Kuss
- Department of Functional Genomics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Ana Tzvetkova
- Department of Functional Genomics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Xiaomin Wang
- Department of Cardiology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 12200 Berlin, Germany
| | - Liliana H Mochmann
- Department of Cardiology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 12200 Berlin, Germany
| | - Pegah Ramezani Rad
- Department of Cardiology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 12200 Berlin, Germany
| | - Stefan Weiss
- Department of Functional Genomics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), Site Greifswald, 17487 Greifswald, Germany
| | - Stefan Simm
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Tanja Zeller
- University Center of Cardiovascular Science, University Heart and Vascular Center, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Site Hamburg/Lübeck/Kiel, 20246 Hamburg, Germany
| | - Henry Voelzke
- German Center for Cardiovascular Research (DZHK), Site Greifswald, 17487 Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Wolfgang Hoffmann
- German Center for Cardiovascular Research (DZHK), Site Greifswald, 17487 Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), Site Greifswald, 17487 Greifswald, Germany
| | - Stefan B Felix
- German Center for Cardiovascular Research (DZHK), Site Greifswald, 17487 Greifswald, Germany
- Department of Cardiology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Marcus Dörr
- German Center for Cardiovascular Research (DZHK), Site Greifswald, 17487 Greifswald, Germany
- Department of Cardiology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Antje Beling
- German Center for Cardiovascular Research (DZHK), Site Berlin, 12200 Berlin, Germany
- Institute for Biochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Carsten Skurk
- Department of Cardiology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 12200 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, 12200 Berlin, Germany
| | - David-Manuel Leistner
- Department of Cardiology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 12200 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, 12200 Berlin, Germany
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Bernhard H Rauch
- German Center for Cardiovascular Research (DZHK), Site Greifswald, 17487 Greifswald, Germany
- Institute for Pharmacology, University Medicine Greifswald, 17487 Greifswald, Germany
- Department Human Medicine, Section Pharmacology and Toxicology, Carl von Ossietzky Universität, 26129 Oldenburg, Germany
| | - Tetsuro Hirose
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita 565-0871, Japan
| | - Bettina Heidecker
- Department of Cardiology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 12200 Berlin, Germany
| | - Karin Klingel
- Institute for Pathology and Neuropathology, Department of Pathology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Shinichi Nakagawa
- RNA Biology Laboratory, RIKEN Advanced Research Institute, Wako, Saitama 351-0198, Japan
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Wolfram C Poller
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Arash Haghikia
- Department of Cardiology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 12200 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, 12200 Berlin, Germany
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Wolfgang Poller
- Department of Cardiology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 12200 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, 12200 Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany
| |
Collapse
|
11
|
Hu W, Yang C, Guo X, Wu Y, Loh XJ, Li Z, Wu YL, Wu C. Research Advances of Injectable Functional Hydrogel Materials in the Treatment of Myocardial Infarction. Gels 2022; 8:423. [PMID: 35877508 PMCID: PMC9316750 DOI: 10.3390/gels8070423] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/30/2022] [Accepted: 07/03/2022] [Indexed: 12/10/2022] Open
Abstract
Myocardial infarction (MI) has become one of the serious diseases threatening human life and health. However, traditional treatment methods for MI have some limitations, such as irreversible myocardial necrosis and cardiac dysfunction. Fortunately, recent endeavors have shown that hydrogel materials can effectively prevent negative remodeling of the heart and improve the heart function and long-term prognosis of patients with MI due to their good biocompatibility, mechanical properties, and electrical conductivity. Therefore, this review aims to summarize the research progress of injectable hydrogel in the treatment of MI in recent years and to introduce the rational design of injectable hydrogels in myocardial repair. Finally, the potential challenges and perspectives of injectable hydrogel in this field will be discussed, in order to provide theoretical guidance for the development of new and effective treatment strategies for MI.
Collapse
Affiliation(s)
- Wei Hu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Cui Yang
- School of Medicine, Xiamen University, Xiamen 361003, China;
| | - Xiaodan Guo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Yihong Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore;
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore;
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE) Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| |
Collapse
|
12
|
Luraghi A, Ferrandi M, Barassi P, Arici M, Hsu SC, Torre E, Ronchi C, Romerio A, Chang GJ, Ferrari P, Bianchi G, Zaza A, Rocchetti M, Peri F. Highly Selective SERCA2a Activators: Preclinical Development of a Congeneric Group of First-in-Class Drug Leads against Heart Failure. J Med Chem 2022; 65:7324-7333. [PMID: 35580334 PMCID: PMC9150102 DOI: 10.1021/acs.jmedchem.2c00347] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Indexed: 11/30/2022]
Abstract
The stimulation of sarcoplasmic reticulum calcium ATPase SERCA2a emerged as a novel therapeutic strategy to efficiently improve overall cardiac function in heart failure (HF) with reduced arrhythmogenic risk. Istaroxime is a clinical-phase IIb compound with a double mechanism of action, Na+/K+ ATPase inhibition and SERCA2a stimulation. Starting from the observation that istaroxime metabolite PST3093 does not inhibit Na+/K+ ATPase while stimulates SERCA2a, we synthesized a series of bioisosteric PST3093 analogues devoid of Na+/K+ ATPase inhibitory activity. Most of them retained SERCA2a stimulatory action with nanomolar potency in cardiac preparations from healthy guinea pigs and streptozotocin (STZ)-treated rats. One compound was further characterized in isolated cardiomyocytes, confirming SERCA2a stimulation and in vivo showing a safety profile and improvement of cardiac performance following acute infusion in STZ rats. We identified a new class of selective SERCA2a activators as first-in-class drug candidates for HF treatment.
Collapse
Affiliation(s)
- Andrea Luraghi
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Milano 20126, Italy
| | - Mara Ferrandi
- Windtree
Therapeutics Inc., Warrington, Pennsylvania 18976, United States
| | - Paolo Barassi
- Windtree
Therapeutics Inc., Warrington, Pennsylvania 18976, United States
| | - Martina Arici
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Milano 20126, Italy
| | | | - Eleonora Torre
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Milano 20126, Italy
| | - Carlotta Ronchi
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Milano 20126, Italy
| | - Alessio Romerio
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Milano 20126, Italy
| | - Gwo-Jyh Chang
- Cardiovascular
Medicine, Chang Gung University, Tao-Yuan 333323 Taiwan
| | - Patrizia Ferrari
- Windtree
Therapeutics Inc., Warrington, Pennsylvania 18976, United States
| | - Giuseppe Bianchi
- Windtree
Therapeutics Inc., Warrington, Pennsylvania 18976, United States
- Università
Vita-Salute San Raffaele, Milano 20132, Italy
| | - Antonio Zaza
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Milano 20126, Italy
| | - Marcella Rocchetti
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Milano 20126, Italy
| | - Francesco Peri
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Milano 20126, Italy
| |
Collapse
|
13
|
Tabasso C, Frossard MP, Ducret C, Chehade H, Mauduit C, Benahmed M, Simeoni U, Siddeek B. Transient Post-Natal Exposure to Xenoestrogens Induces Long-Term Alterations in Cardiac Calcium Signaling. TOXICS 2022; 10:toxics10030102. [PMID: 35324727 PMCID: PMC8954167 DOI: 10.3390/toxics10030102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/03/2022] [Accepted: 02/16/2022] [Indexed: 02/05/2023]
Abstract
Today, non-communicable disorders are widespread worldwide. Among them, cardiovascular diseases represent the main cause of death. At the origin of these diseases, exposure to challenges during developmental windows of vulnerability (peri-conception, in utero, and early infancy periods) have been incriminated. Among the challenges that have been described, endocrine disruptors are of high concern because of their omnipresence in the environment. Worrisomely, since birth, children are exposed to a significant number of endocrine disruptors. However, the role of such early exposure on long-term cardiac health is poorly described. In this context, based on a model of rats exposed postnatally and transiently to an estrogenic compound prototype (estradiol benzoate, EB), we aimed to delineate the effects on the adult heart of such transient early exposure to endocrine disruptors and identify the underlying mechanisms involved in the potential pathogenesis. We found that this transient post-natal exposure to EB induced cardiac hypertrophy in adulthood, with increased cardiomyocyte size. The evaluation of cardiac calcium signaling, through immunoblot approaches, highlighted decreased expression of the sarcoplasmic reticulum calcium ATPase 2 (SERCA2) and decreased Nuclear Factor of Activated T Cells (NFAT3) phosphorylation as a potential underlying mechanism of cardiac hypertrophy. Furthermore, the treatment of cardiomyocytes with EB in vitro induced a decrease in SERCA2 protein levels. Overall, our study demonstrates that early transient exposure to EB induces permanent cardiac alterations. Together, our data highlight SERCA2 down-regulation as a potential mechanism involved in the cardiac pathogenesis induced by EB. These results suggest programming of adult heart dysfunctions such as arrhythmia and heart failures by early exposure to endocrine disruptors and could open new perspectives for treatment and prevention.
Collapse
Affiliation(s)
- Cassandra Tabasso
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
| | - Marie-Pauline Frossard
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
| | - Camille Ducret
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
| | - Hassib Chehade
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
| | - Claire Mauduit
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 10, 06204 Nice, France; (C.M.); (M.B.)
| | - Mohamed Benahmed
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 10, 06204 Nice, France; (C.M.); (M.B.)
| | - Umberto Simeoni
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
| | - Benazir Siddeek
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
- Correspondence: ; Tel.: +41-21-3143-212
| |
Collapse
|
14
|
Morciano G, Rimessi A, Patergnani S, Vitto VAM, Danese A, Kahsay A, Palumbo L, Bonora M, Wieckowski MR, Giorgi C, Pinton P. Calcium dysregulation in heart diseases: Targeting calcium channels to achieve a correct calcium homeostasis. Pharmacol Res 2022; 177:106119. [PMID: 35131483 DOI: 10.1016/j.phrs.2022.106119] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 12/16/2022]
Abstract
Intracellular calcium signaling is a universal language source shared by the most part of biological entities inside cells that, all together, give rise to physiological and functional anatomical units, the organ. Although preferentially recognized as signaling between cell life and death processes, in the heart it assumes additional relevance considered the importance of calcium cycling coupled to ATP consumption in excitation-contraction coupling. The concerted action of a plethora of exchangers, channels and pumps inward and outward calcium fluxes where needed, to convert energy and electric impulses in muscle contraction. All this without realizing it, thousands of times, every day. An improper function of those proteins (i.e., variation in expression, mutations onset, dysregulated channeling, differential protein-protein interactions) being part of this signaling network triggers a short circuit with severe acute and chronic pathological consequences reported as arrhythmias, cardiac remodeling, heart failure, reperfusion injury and cardiomyopathies. By acting with chemical, peptide-based and pharmacological modulators of these players, a correction of calcium homeostasis can be achieved accompanied by an amelioration of clinical symptoms. This review will focus on all those defects in calcium homeostasis which occur in the most common cardiac diseases, including myocardial infarction, arrhythmia, hypertrophy, heart failure and cardiomyopathies. This part will be introduced by the state of the art on the proteins involved in calcium homeostasis in cardiomyocytes and followed by the therapeutic treatments that to date, are able to target them and to revert the pathological phenotype.
Collapse
Affiliation(s)
- Giampaolo Morciano
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, RA, Italy.
| | - Alessandro Rimessi
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Simone Patergnani
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Veronica A M Vitto
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Alberto Danese
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Asrat Kahsay
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Laura Palumbo
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Massimo Bonora
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism. Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, RA, Italy.
| |
Collapse
|
15
|
Wen ZJ, Xin H, Wang YC, Liu HW, Gao YY, Zhang YF. Emerging roles of circRNAs in the pathological process of myocardial infarction. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:828-848. [PMID: 34729251 PMCID: PMC8536508 DOI: 10.1016/j.omtn.2021.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Myocardial infarction (MI) is defined as cardiomyocyte death in a clinical context consistent with ischemic insult. MI remains one of the leading causes of morbidity and mortality worldwide. Although there are a number of effective clinical methods for the diagnosis and treatment of MI, further investigation of novel biomarkers and molecular therapeutic targets is required. Circular RNAs (circRNAs), novel non-coding RNAs, have been reported to function mainly by acting as microRNA (miRNA) sponges or binding to RNA-binding proteins (RBPs). The circRNA-miRNA-mRNA (protein) regulatory pathway regulates gene expression and affects the pathological mechanisms of various diseases. Undoubtedly, a more comprehensive understanding of the relationship between MI and circRNA will lay the foundation for the development of circRNA-based diagnostic and therapeutic strategies for MI. Therefore, this review summarizes the pathophysiological process of MI and various approaches to measure circRNA levels in MI patients, tissues, and cells; highlights the significance of circRNAs in the regulation MI pathogenesis and development; and provides potential clinical insight for the diagnosis, prognosis, and treatment of MI.
Collapse
Affiliation(s)
- Zeng-Jin Wen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yong-Chen Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Hao-Wen Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yan-Yan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
16
|
Schultheiss HP, Baumeier C, Pietsch H, Bock CT, Poller W, Escher F. Cardiovascular consequences of viral infections: from COVID to other viral diseases. Cardiovasc Res 2021; 117:2610-2623. [PMID: 34609508 PMCID: PMC8500164 DOI: 10.1093/cvr/cvab315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022] Open
Abstract
Infection of the heart muscle with cardiotropic viruses is one of the major aetiologies of myocarditis and acute and chronic inflammatory cardiomyopathy (DCMi). However, viral myocarditis and subsequent dilated cardiomyopathy is still a challenging disease to diagnose and to treat and is therefore a significant public health issue globally. Advances in clinical examination and thorough molecular genetic analysis of intramyocardial viruses and their activation status have incrementally improved our understanding of molecular pathogenesis and pathophysiology of viral infections of the heart muscle. To date, several cardiotropic viruses have been implicated as causes of myocarditis and DCMi. These include, among others, classical cardiotropic enteroviruses (Coxsackieviruses B), the most commonly detected parvovirus B19, and human herpes virus 6. A newcomer is the respiratory virus that has triggered the worst pandemic in a century, SARS-CoV-2, whose involvement and impact in viral cardiovascular disease is under scrutiny. Despite extensive research into the pathomechanisms of viral infections of the cardiovascular system, our knowledge regarding their treatment and management is still incomplete. Accordingly, in this review, we aim to explore and summarize the current knowledge and available evidence on viral infections of the heart. We focus on diagnostics, clinical relevance and cardiovascular consequences, pathophysiology, and current and novel treatment strategies.
Collapse
Affiliation(s)
| | - Christian Baumeier
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, Berlin, Germany
| | - Heiko Pietsch
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, Berlin, Germany
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - C -Thomas Bock
- Division of Viral Gastroenteritis and Hepatitis Pathogens and Enteroviruses, Department of Infectious Diseases, Robert Koch Institute, Berlin 13353 Germany
- Institute of Tropical Medicine, University of Tübingen, Tübingen 72074, Germany
| | - Wolfgang Poller
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Department of Cardiology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin12203, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany
| | - Felicitas Escher
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, Berlin, Germany
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
17
|
Oxidized LDL but not angiotensin II induces cardiomyocyte hypertrophic responses through the interaction between LOX-1 and AT 1 receptors. J Mol Cell Cardiol 2021; 162:110-118. [PMID: 34555408 DOI: 10.1016/j.yjmcc.2021.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/15/2021] [Accepted: 09/08/2021] [Indexed: 01/19/2023]
Abstract
It is well known that lectin-like oxidized low-density lipoprotein (ox-LDL) and its receptor LOX-1, angiotensin II (AngII) and its type 1 receptor (AT1-R) play an important role in the development of cardiac hypertrophy. However, the molecular mechanism is not clear. In this study, we found that ox-LDL-induced cardiac hypertrophy was suppressed by inhibition of LOX-1 or AT1-R but not by AngII inhibition. These results suggest that the receptors LOX-1 and AT1-R, rather than AngII, play a key role in the role of ox-LDL. The same results were obtained in mice lacking endogenous AngII and their isolated cardiomyocytes. Ox-LDL but not AngII could induce the binding of LOX-1 and AT1-R; inhibition of LOX-1 or AT1-R but not AngII could abolish the binding of these two receptors. Overexpression of wild type LOX-1 with AT1-R enhanced ox-LDL-induced binding of two receptors and phosphorylation of ERKs, however, transfection of LOX-1 dominant negative mutant (lys266ala / lys267ala) or an AT1-R mutant (glu257ala) not only reduced the binding of two receptors but also inhibited the ERKs phosphorylation. Phosphorylation of ERKs induced by ox-LDL in LOX-1 and AT1-R-overexpression cells was abrogated by an inhibitor of Gq protein rather than Jak2, Rac1 or RhoA. Genetically, an AT1-R mutant lacking Gq protein coupling ability inhibited ox-LDL induced ERKs phosphorylation. Furthermore, through bimolecular fluorescence complementation analysis, we confirmed that ox-LDL rather than AngII stimulation induced the direct binding of LOX-1 and AT1-R. We conclude that direct binding of LOX-1 and AT1-R and the activation of downstream Gq protein are important mechanisms of ox-LDL-induced cardiomyocyte hypertrophy.
Collapse
|
18
|
Landmesser U, Poller W, Tsimikas S, Most P, Paneni F, Lüscher TF. From traditional pharmacological towards nucleic acid-based therapies for cardiovascular diseases. Eur Heart J 2021; 41:3884-3899. [PMID: 32350510 DOI: 10.1093/eurheartj/ehaa229] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/17/2020] [Accepted: 03/12/2020] [Indexed: 02/06/2023] Open
Abstract
Nucleic acid-based therapeutics are currently developed at large scale for prevention and management of cardiovascular diseases (CVDs), since: (i) genetic studies have highlighted novel therapeutic targets suggested to be causal for CVD; (ii) there is a substantial recent progress in delivery, efficacy, and safety of nucleic acid-based therapies; (iii) they enable effective modulation of therapeutic targets that cannot be sufficiently or optimally addressed using traditional small molecule drugs or antibodies. Nucleic acid-based therapeutics include (i) RNA-targeted therapeutics for gene silencing; (ii) microRNA-modulating and epigenetic therapies; (iii) gene therapies; and (iv) genome-editing approaches (e.g. CRISPR-Cas-based): (i) RNA-targeted therapeutics: several large-scale clinical development programmes, using antisense oligonucleotides (ASO) or short interfering RNA (siRNA) therapeutics for prevention and management of CVD have been initiated. These include ASO and/or siRNA molecules to lower apolipoprotein (a) [apo(a)], proprotein convertase subtilisin/kexin type 9 (PCSK9), apoCIII, ANGPTL3, or transthyretin (TTR) for prevention and treatment of patients with atherosclerotic CVD or TTR amyloidosis. (ii) MicroRNA-modulating and epigenetic therapies: novel potential therapeutic targets are continually arising from human non-coding genome and epigenetic research. First microRNA-based therapeutics or therapies targeting epigenetic regulatory pathways are in clinical studies. (iii) Gene therapies: EMA/FDA have approved gene therapies for non-cardiac monogenic diseases and LDL receptor gene therapy is currently being examined in patients with homozygous hypercholesterolaemia. In experimental studies, gene therapy has significantly improved cardiac function in heart failure animal models. (iv) Genome editing approaches: these technologies, such as using CRISPR-Cas, have proven powerful in stem cells, however, important challenges are remaining, e.g. low rates of homology-directed repair in somatic cells such as cardiomyocytes. In summary, RNA-targeted therapies (e.g. apo(a)-ASO and PCSK9-siRNA) are now in large-scale clinical outcome trials and will most likely become a novel effective and safe therapeutic option for CVD in the near future. MicroRNA-modulating, epigenetic, and gene therapies are tested in early clinical studies for CVD. CRISPR-Cas-mediated genome editing is highly effective in stem cells, but major challenges are remaining in somatic cells, however, this field is rapidly advancing.
Collapse
Affiliation(s)
- Ulf Landmesser
- Department of Cardiology, Campus Benjamin Franklin, CC11 (Cardiovascular Medicine), Charite-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health, Anna-Louisa-Karsch-Strasse 2, 10178 Berlin, Germany
| | - Wolfgang Poller
- Department of Cardiology, Campus Benjamin Franklin, CC11 (Cardiovascular Medicine), Charite-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, 9500 Gilman Drive, BSB 1080, La Jolla, CA 92093-0682, USA
| | - Patrick Most
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany.,Center for Translational Medicine, Jefferson Medical College, 1020 Locust Street, Philadelphia, PA 19107, USA.,Molecular and Translational Cardiology, Department of Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 669, 69120 Heidelberg, Germany
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Rämistrasse 100, MOU2, 8091 Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland.,Research, Education and Development, Royal Brompton and Harefield Hospital Trust and Imperial College London, National Heart and Lung Institute, Guy Scadding Building, Dovehouse Street, London SW3 6LY, UK
| |
Collapse
|
19
|
Torre E, Arici M, Lodrini AM, Ferrandi M, Barassi P, Hsu SC, Chang GJ, Boz E, Sala E, Vagni S, Altomare C, Mostacciuolo G, Bussadori C, Ferrari P, Bianchi G, Rocchetti M. SERCA2a stimulation by istaroxime improves intracellular Ca2+ handling and diastolic dysfunction in a model of diabetic cardiomyopathy. Cardiovasc Res 2021; 118:1020-1032. [PMID: 33792692 PMCID: PMC8930067 DOI: 10.1093/cvr/cvab123] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/20/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022] Open
Abstract
Aims Diabetic cardiomyopathy is a multifactorial disease characterized by an early onset of diastolic dysfunction (DD) that precedes the development of systolic impairment. Mechanisms that can restore cardiac relaxation improving intracellular Ca2+ dynamics represent a promising therapeutic approach for cardiovascular diseases associated to DD. Istaroxime has the dual properties to accelerate Ca2+ uptake into sarcoplasmic reticulum (SR) through the SR Ca2+ pump (SERCA2a) stimulation and to inhibit Na+/K+ ATPase (NKA). This project aims to characterize istaroxime effects at a concentration (100 nmol/L) marginally affecting NKA, in order to highlight its effects dependent on the stimulation of SERCA2a in an animal model of mild diabetes. Methods and results Streptozotocin (STZ) treated diabetic rats were studied at 9 weeks after STZ injection in comparison to controls (CTR). Istaroxime effects were evaluated in vivo and in left ventricular (LV) preparations. STZ animals showed (i) marked DD not associated to cardiac fibrosis, (ii) LV mass reduction associated to reduced LV cell dimension and T-tubules loss, (iii) reduced LV SERCA2 protein level and activity and (iv) slower SR Ca2+ uptake rate, (v) LV action potential (AP) prolongation and increased short-term variability (STV) of AP duration, (vi) increased diastolic Ca2+, and (vii) unaltered SR Ca2+ content and stability in intact cells. Acute istaroxime infusion (0.11 mg/kg/min for 15 min) reduced DD in STZ rats. Accordingly, in STZ myocytes istaroxime (100 nmol/L) stimulated SERCA2a activity and blunted STZ-induced abnormalities in LV Ca2+ dynamics. In CTR myocytes, istaroxime increased diastolic Ca2+ level due to NKA blockade albeit minimal, while its effects on SERCA2a were almost absent. Conclusions SERCA2a stimulation by istaroxime improved STZ-induced DD and intracellular Ca2+ handling anomalies. Thus, SERCA2a stimulation can be considered a promising therapeutic approach for DD treatment.
Collapse
Affiliation(s)
- Eleonora Torre
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Martina Arici
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Alessandra Maria Lodrini
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Mara Ferrandi
- Windtree Therapeutics Inc., Warrington, Pennsylvania, USA
| | - Paolo Barassi
- Windtree Therapeutics Inc., Warrington, Pennsylvania, USA
| | | | | | | | - Emanuela Sala
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Sara Vagni
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | | | - Gaspare Mostacciuolo
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | | | | | | | - Marcella Rocchetti
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| |
Collapse
|
20
|
Rilo-Alvarez H, Ledo AM, Vidal A, Garcia-Fuentes M. Delivery of transcription factors as modulators of cell differentiation. Drug Deliv Transl Res 2021; 11:426-444. [PMID: 33611769 DOI: 10.1007/s13346-021-00931-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 12/13/2022]
Abstract
Fundamental studies performed during the last decades have shown that cell fate is much more plastic than previously considered, and technologies for its manipulation are a keystone for many new tissue regeneration therapies. Transcription factors (TFs) are DNA-binding proteins that control gene expression, and they have critical roles in the control of cell fate and other cellular behavior. TF-based therapies have much medical potential, but their use as drugs depends on the development of suitable delivery technologies that can help them reach their action site inside of the cells. TFs can be used either as proteins or encoded in polynucleotides. When used in protein form, many TFs require to be associated to a cell-penetrating peptide or another transduction domain. As polynucleotides, they can be delivered either by viral carriers or by non-viral systems such as polyplexes and lipoplexes. TF-based therapies have extensively shown their potential to solve many tissue-engineering problems, including bone, cartilage and cardiac regeneration. Yet, their use has expanded beyond regenerative medicine to other prominent disease areas such as cancer therapy and immunomodulation. This review summarizes some of the delivery options for effective TF-based therapies and their current main applications.
Collapse
Affiliation(s)
- Héctor Rilo-Alvarez
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Adriana M Ledo
- Respiratory Therapeutic Area, Novartis Institutes for BioMedical Research, Inc, 700 Main Street, Cambridge, MA, 02139, USA
| | - Anxo Vidal
- Department of Physiology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Marcos Garcia-Fuentes
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| |
Collapse
|
21
|
Anger M, Scheufele F, Ramanujam D, Meyer K, Nakajima H, Field LJ, Engelhardt S, Sarikas A. Genetic ablation of Cullin-RING E3 ubiquitin ligase 7 restrains pressure overload-induced myocardial fibrosis. PLoS One 2020; 15:e0244096. [PMID: 33351822 PMCID: PMC7755222 DOI: 10.1371/journal.pone.0244096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/03/2020] [Indexed: 11/20/2022] Open
Abstract
Fibrosis is a pathognomonic feature of structural heart disease and counteracted by distinct cardioprotective mechanisms, e.g. activation of the phosphoinositide 3-kinase (PI3K) / AKT pro-survival pathway. The Cullin-RING E3 ubiquitin ligase 7 (CRL7) was identified as negative regulator of PI3K/AKT signalling in skeletal muscle, but its role in the heart remains to be elucidated. Here, we sought to determine whether CRL7 modulates to cardiac fibrosis following pressure overload and dissect its underlying mechanisms. For inactivation of CRL7, the Cullin 7 (Cul7) gene was deleted in cardiac myocytes (CM) by injection of adeno-associated virus subtype 9 (AAV9) vectors encoding codon improved Cre-recombinase (AAV9-CMV-iCre) in Cul7flox/flox mice. In addition, Myosin Heavy Chain 6 (Myh6; alpha-MHC)-MerCreMer transgenic mice with tamoxifen-induced CM-specific expression of iCre were used as alternate model. After transverse aortic constriction (TAC), causing chronic pressure overload and fibrosis, AAV9-CMV-iCre induced Cul7-/- mice displayed a ~50% reduction of interstitial cardiac fibrosis when compared to Cul7+/+ animals (6.7% vs. 3.4%, p<0.01). Similar results were obtained with Cul7flox/floxMyh6-Mer-Cre-MerTg(1/0) mice which displayed a ~30% reduction of cardiac fibrosis after TAC when compared to Cul7+/+Myh6-Mer-Cre-MerTg(1/0) controls after TAC surgery (12.4% vs. 8.7%, p<0.05). No hemodynamic alterations were observed. AKTSer473 phosphorylation was increased 3-fold (p<0.01) in Cul7-/- vs. control mice, together with a ~78% (p<0.001) reduction of TUNEL-positive apoptotic cells three weeks after TAC. In addition, CM-specific expression of a dominant-negative CUL71152stop mutant resulted in a 16.3-fold decrease (p<0.001) of in situ end-labelling (ISEL) positive apoptotic cells. Collectively, our data demonstrate that CM-specific ablation of Cul7 restrains myocardial fibrosis and apoptosis upon pressure overload, and introduce CRL7 as a potential target for anti-fibrotic therapeutic strategies of the heart.
Collapse
Affiliation(s)
- Melanie Anger
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Florian Scheufele
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Deepak Ramanujam
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Kathleen Meyer
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Hidehiro Nakajima
- Wells Center for Pediatric Research and Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Loren J. Field
- Wells Center for Pediatric Research and Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Antonio Sarikas
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
- * E-mail:
| |
Collapse
|
22
|
Sun J, Niu C, Ye W, An N, Chen G, Huang X, Wang J, Chen X, Shen Y, Huang S, Wang Y, Wang X, Wang Y, Jin L, Cong W, Li X. FGF13 Is a Novel Regulator of NF-κB and Potentiates Pathological Cardiac Hypertrophy. iScience 2020; 23:101627. [PMID: 33089113 PMCID: PMC7567043 DOI: 10.1016/j.isci.2020.101627] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 08/13/2020] [Accepted: 09/25/2020] [Indexed: 12/31/2022] Open
Abstract
FGF13 is an intracellular FGF factor. Its role in cardiomyopathies has been rarely investigated. We revealed that endogenous FGF13 is up-regulated in cardiac hypertrophy accompanied by increased nuclear localization. The upregulation of FGF13 plays a deteriorating role both in hypertrophic cardiomyocytes and mouse hearts. Mechanistically, FGF13 directly interacts with p65 by its nuclear localization sequence and co-localizes with p65 in the nucleus in cardiac hypertrophy. FGF13 deficiency inhibits NF-κB activation in ISO-treated NRCMs and TAC-surgery mouse hearts, whereas FGF13 overexpression shows the opposite trend. Moreover, FGF13 overexpression alone is sufficient to activate NF-κB in cardiomyocytes. The interaction between FGF13 and p65 or the effects of FGF13 on NF-κB have nothing to do with IκB. Together, an IκB-independent mechanism for NF-κB regulation has been revealed in cardiomyocytes both under basal and stressful conditions, suggesting the promising application of FGF13 as a therapeutic target for pathological cardiac hypertrophy and heart failure. Endogenous FGF13 is up-regulated in cardiomyocytes under pressure overload FGF13 directly interacts with p65 Forced FGF13 overexpression activates NF-κB in cardiomyocytes
Collapse
Affiliation(s)
- Jia Sun
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Chao Niu
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Weijian Ye
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ning An
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Gen Chen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaozhong Huang
- Department of Pediatric Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jianan Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Xixi Chen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Yingjie Shen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Shuai Huang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Ying Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Xu Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Yang Wang
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University Wenzhou, 325000, China
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
23
|
García-Manzanares M, Tarazón E, Ortega A, Gil-Cayuela C, Martínez-Dolz L, González-Juanatey JR, Lago F, Portolés M, Roselló-Lletí E, Rivera M. XPO1 Gene Therapy Attenuates Cardiac Dysfunction in Rats with Chronic Induced Myocardial Infarction. J Cardiovasc Transl Res 2020; 13:593-600. [PMID: 31768947 PMCID: PMC7423868 DOI: 10.1007/s12265-019-09932-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/11/2019] [Indexed: 10/25/2022]
Abstract
Transcriptomic signature of XPO1 was highly expressed and inversely related to left ventricular function in ischemic cardiomyopathy patients. We hypothesized that treatment with AAV9-shXPO1 attenuates left ventricular dysfunction and remodeling in a myocardial infarction rat model. We induced myocardial infarction by coronary ligation in Sprague-Dawley rats (n = 10), which received AAV9-shXPO1 (n = 5) or placebo AAV9-scramble (n = 5) treatment. Serial echocardiographic assessment was performed throughout the study. After myocardial infarction, AAV9-shXPO1-treated rats showed partial recovery of left ventricular fractional shortening (16.8 ± 2.8 vs 24.6 ± 4.1%, P < 0.05) and a maintained left ventricular dimension (6.17 ± 0.95 vs 4.70 ± 0.93 mm, P < 0.05), which was not observed in non-treated rats. Furthermore, lower levels of EXP-1 (P < 0.05) and lower collagen fibers and fibrosis in cardiac tissue were observed. However, no differences were found in the IL-6 or TNFR1 plasma levels of the myocardium of AAV9-shXPO1 rats. AAV9-shXPO1 administration attenuates cardiac dysfunction and remodeling in rats after myocardial infarction, producing the gene silencing of XPO1.
Collapse
Affiliation(s)
- María García-Manzanares
- Department of Animal Medicine and Surgery, Veterinary Faculty, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Estefanía Tarazón
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Ana Ortega
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Carolina Gil-Cayuela
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Luis Martínez-Dolz
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
- Heart Failure and Transplantation Unit, Cardiology Department, University Hospital La Fe, Valencia, Spain
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Manuel Portolés
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - Esther Roselló-Lletí
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Veterinary Faculty, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Miguel Rivera
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| |
Collapse
|
24
|
Song P, Zhao Q, Zou MH. Targeting senescent cells to attenuate cardiovascular disease progression. Ageing Res Rev 2020; 60:101072. [PMID: 32298812 DOI: 10.1016/j.arr.2020.101072] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD) is the most common disease to increase as life expectancy increases. Most high-profile pharmacological treatments for age-related CVD have led to inefficacious results, implying that novel approaches to treating these pathologies are needed. Emerging data have demonstrated that senescent cardiovascular cells, which are characterized by irreversible cell cycle arrest and a distinct senescence-associated secretory phenotype, accumulate in aged or diseased cardiovascular systems, suggesting that they may impair cardiovascular function. This review discusses the evidence implicating senescent cells in cardiovascular ageing, the onset and progression of CVD, and the molecular mechanisms underlying cardiovascular cell senescence. We also review eradication of senescent cardiovascular cells by small-molecule-drug-mediated apoptosis and immune cell-mediated efferocytosis and toxicity as promising and precisely targeted therapeutics for CVD prevention and treatment.
Collapse
|
25
|
Sadat-Ebrahimi SR, Aslanabadi N. Role of MicroRNAs in Diagnosis, Prognosis, and Treatment of Acute Heart Failure: Ambassadors from Intracellular Zone. Galen Med J 2020; 9:e1818. [PMID: 34466598 PMCID: PMC8343948 DOI: 10.31661/gmj.v9i0.1818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/24/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
Acute heart failure (AHF) is one of the burdensome diseases affecting a considerable proportion of the population. Recently, it has been demonstrated that micro-ribonucleic acids (miRNAs) can exert diagnostic, prognostic, and therapeutic roles in a variety of conditions including AHF. These molecules play essential roles in HF-related pathophysiology, particularly, cardiac fibrosis, and hypertrophy. Some miRNAs namely miRNA-423-5p are reported to have both diagnostic and prognostic capabilities. However, some studies suggest that combination of biomarkers is a much better way to achieve the highest accuracy such as the combination of miRNAs and N-terminal pro b-type Natriuretic Peptide (NT pro-BNP). Therefore, this review discusses different views towards various roles of miRNAs in AHF.
Collapse
Affiliation(s)
- Seyyed-Reza Sadat-Ebrahimi
- Cardiovascular Research Center, Madani Heart Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naser Aslanabadi
- Cardiovascular Research Center, Madani Heart Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Correspondence to: Naser Aslanabadi, Professor of Cardiology, Cardiovascular Research Center, Madani Heart Center, Tabriz University of Medical Sciences, Tabriz, Iran Telephone Number: +989143110844 Email Address:
| |
Collapse
|
26
|
Xu T, Zhang K, Kan F, Li F, Yu B, Du W, Nie H. Adeno-associated Virus 9-mediated Small RNA Interference of TLR4 Alleviates Myocardial Ischemia and Reperfusion Injury by Inhibition of the NF-κB and MAPK Signaling Pathways in Rats. Curr Mol Med 2020; 19:127-135. [PMID: 30854968 DOI: 10.2174/1566524019666190311122521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND Despite intensive investigation, effective therapeutic procedures for myocardial I/R injury are still in demand. OBJECTIVE To explore the effect of adeno-associated virus 9 (AAV9)-mediated small interfering RNA targeting TLR4 in the treatment of myocardial ischemia and reperfusion (I/R) injury and its influence on the NF-κB and MAPK signaling pathways. METHODS Rats were divided into 3 groups, namely, the sham, AAV9-siRNA control, and AAV9-TLR4 siRNA groups. siRNA solution or normal saline was injected through the tail vain. The rat myocardial I/R injury model was then established. HE staining and TUNEL staining were applied to compare the pathological changes in cardiomyocytes in the three groups. Immunohistochemical staining and western blotting were utilized to detect TLR4 expression under siRNA interference. Serum inflammatory factor (IL-1β, TNF-α) expression was determined by an ELISA commercial kit. Key proteins in the MAPK (p38, JNK 1/2) and NF-κB (p65) signaling pathways were determined to identify the TLR4 siRNA functional mechanism. RESULTS Fluorescence microscopic images of the myocardium indicated that AAV9- mediated siRNA was efficiently transfected into the myocardium, and the infarcted size after I/R injury was decreased by AAV9-TLR4 siRNA when compared with negative control rats (P<0.05). TLR4 protein expression was significantly decreased by siRNA interference (P<0.001). Apoptosis-related factor BCL-2 expression was increased in the TLR4 gene silencing group, whereas Bax expression was decreased. The Bax/BCL-2 ratio was also decreased, demonstrating a protective effect for cardiomyocytes. Inflammatory factors were lower in the TLR4 gene silencing group than in the siRNA control group (P<0.001). The MAPK and NF-κB signaling pathways were activated in myocardial I/R injury; however, the primary proteins in these two signaling pathways were downregulated upon interference of TLR4 siRNA, with significant differences (P<0.05). CONCLUSION AAV9-TLR4 siRNA has a positive effect on myocardial I/R injury by inhibiting the MAPK and NF-κB signaling pathways and can be used as a potential therapeutic method for myocardial I/R injury.
Collapse
Affiliation(s)
- Ting Xu
- Harbin Medical University, Harbin, China
| | - Kuikui Zhang
- Department of Cardiology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | | | - Fengqin Li
- Harbin Medical University, Harbin, China
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjuan Du
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Honggang Nie
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
27
|
Deng Y, Zhang X, Shen H, He Q, Wu Z, Liao W, Yuan M. Application of the Nano-Drug Delivery System in Treatment of Cardiovascular Diseases. Front Bioeng Biotechnol 2020; 7:489. [PMID: 32083068 PMCID: PMC7005934 DOI: 10.3389/fbioe.2019.00489] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/31/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVDs) have become a serious threat to human life and health. Though many drugs acting via different mechanism of action are available in the market as conventional formulations for the treatment of CVDs, they are still far from satisfactory due to poor water solubility, low biological efficacy, non-targeting, and drug resistance. Nano-drug delivery systems (NDDSs) provide a new drug delivery method for the treatment of CVDs with the development of nanotechnology, demonstrating great advantages in solving the above problems. Nevertheless, there are some problems about NDDSs need to be addressed, such as cytotoxicity. In this review, the types and targeting strategies of NDDSs were summarized, and the new research progress in the diagnosis and therapy of CVDs in recent years was reviewed. Future prospective for nano-carriers in drug delivery for CVDs includes gene therapy, in order to provide more ideas for the improvement of cardiovascular drugs. In addition, its safety was also discussed in the review.
Collapse
Affiliation(s)
- Yudi Deng
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xudong Zhang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Haibin Shen
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qiangnan He
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zijian Wu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenzhen Liao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Miaomiao Yuan
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
28
|
García‐Olloqui P, Rodriguez‐Madoz JR, Di Scala M, Abizanda G, Vales Á, Olagüe C, Iglesias‐García O, Larequi E, Aguado‐Alvaro LP, Ruiz‐Villalba A, Prosper F, Gonzalez‐Aseguinolaza G, Pelacho B. Effect of heart ischemia and administration route on biodistribution and transduction efficiency of AAV9 vectors. J Tissue Eng Regen Med 2019; 14:123-134. [DOI: 10.1002/term.2974] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/04/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Paula García‐Olloqui
- Regenerative Medicine DepartmentCenter for Applied Medical Research, University of Navarra Pamplona Spain
| | | | - Marianna Di Scala
- Gene Therapy DepartmentFoundation for Applied Medical Research Pamplona Spain
| | - Gloria Abizanda
- Regenerative Medicine DepartmentCenter for Applied Medical Research, University of Navarra Pamplona Spain
| | - África Vales
- Gene Therapy DepartmentFoundation for Applied Medical Research Pamplona Spain
| | - Cristina Olagüe
- Gene Therapy DepartmentFoundation for Applied Medical Research Pamplona Spain
| | - Olalla Iglesias‐García
- Regenerative Medicine DepartmentCenter for Applied Medical Research, University of Navarra Pamplona Spain
| | - Eduardo Larequi
- Regenerative Medicine DepartmentCenter for Applied Medical Research, University of Navarra Pamplona Spain
| | - Laura Pilar Aguado‐Alvaro
- Regenerative Medicine DepartmentCenter for Applied Medical Research, University of Navarra Pamplona Spain
| | - Adrián Ruiz‐Villalba
- Regenerative Medicine DepartmentCenter for Applied Medical Research, University of Navarra Pamplona Spain
| | - Felipe Prosper
- Regenerative Medicine DepartmentCenter for Applied Medical Research, University of Navarra Pamplona Spain
- Hematology and Cell Therapy DepartmentClínica Universidad de Navarra, University of Navarra Pamplona Spain
| | | | - Beatriz Pelacho
- Regenerative Medicine DepartmentCenter for Applied Medical Research, University of Navarra Pamplona Spain
| |
Collapse
|
29
|
Wang XT, Wu XD, Lu YX, Sun YH, Zhu HH, Liang JB, He WK, Li L. Egr-1 is involved in coronary microembolization-induced myocardial injury via Bim/Beclin-1 pathway-mediated autophagy inhibition and apoptosis activation. Aging (Albany NY) 2019; 10:3136-3147. [PMID: 30391937 PMCID: PMC6286823 DOI: 10.18632/aging.101616] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/19/2018] [Indexed: 12/22/2022]
Abstract
Coronary microembolization (CME) substantially reduces the clinical benefits of myocardial reperfusion therapy. Autophagy and apoptosis participate in the pathophysiological processes of almost all cardiovascular diseases, including CME-induced myocardial injury, but the precise underlying mechanisms remain unclear. In the present study, we observed that Egr-1 expression was substantially increased after CME modeling. Inhibition of Egr-1 expression through the targeted delivery of rAAV9-Egr-1-shRNA improved cardiac function and reduced myocardial injury. The microinfarct size was also significantly smaller in the Egr-1 inhibitor group than in the CME group. These benefits were partially reversed by the autophagy inhibitor 3-MA. As shown in our previous study, autophagy in the myocardium was impaired after CME. Inhibition of Egr-1 expression in vivo restored the autophagy flux and reduced myocardial apoptosis, at least partially, by inhibiting the Egr-1/Bim/Beclin-1 pathway, as evidenced by the results of the western blot, RT-qPCR, and TUNEL staining. At the same time, TEM showed a dramatic increase in the number of typical autophagic vacuoles in the Egr-1 inhibitor group compared to the CME group. Based on these findings, the Egr-1/Bim/Beclin-1 pathway may be involved in CME-induced myocardial injury by regulating myocardial autophagy and apoptosis, and this pathway represents a potential therapeutic target in CME.
Collapse
Affiliation(s)
- Xian-Tao Wang
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Xiao-Dan Wu
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yuan-Xi Lu
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yu-Han Sun
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Han-Hua Zhu
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jia-Bao Liang
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Wen-Kai He
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Lang Li
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
30
|
Affiliation(s)
- Dai Kusumoto
- From the Department of Cardiology (D.K., S.Y., K.F.), Keio University School of Medicine, Tokyo, Japan.,Department of Emergency and Critical Care Medicine (D.K.), Keio University School of Medicine, Tokyo, Japan
| | - Shinsuke Yuasa
- From the Department of Cardiology (D.K., S.Y., K.F.), Keio University School of Medicine, Tokyo, Japan
| | - Keiichi Fukuda
- From the Department of Cardiology (D.K., S.Y., K.F.), Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
31
|
Pathophysiology of Calcium Mediated Ventricular Arrhythmias and Novel Therapeutic Options with Focus on Gene Therapy. Int J Mol Sci 2019; 20:ijms20215304. [PMID: 31653119 PMCID: PMC6862059 DOI: 10.3390/ijms20215304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias constitute a major health problem with a huge impact on mortality rates and health care costs. Despite ongoing research efforts, the understanding of the molecular mechanisms and processes responsible for arrhythmogenesis remains incomplete. Given the crucial role of Ca2+-handling in action potential generation and cardiac contraction, Ca2+ channels and Ca2+ handling proteins represent promising targets for suppression of ventricular arrhythmias. Accordingly, we report the different roles of Ca2+-handling in the development of congenital as well as acquired ventricular arrhythmia syndromes. We highlight the therapeutic potential of gene therapy as a novel and innovative approach for future arrhythmia therapy. Furthermore, we discuss various promising cellular and mitochondrial targets for therapeutic gene transfer currently under investigation.
Collapse
|
32
|
Zaleta-Rivera K, Dainis A, Ribeiro AJS, Cordero P, Rubio G, Shang C, Liu J, Finsterbach T, Parikh VN, Sutton S, Seo K, Sinha N, Jain N, Huang Y, Hajjar RJ, Kay MA, Szczesna-Cordary D, Pruitt BL, Wheeler MT, Ashley EA. Allele-Specific Silencing Ameliorates Restrictive Cardiomyopathy Attributable to a Human Myosin Regulatory Light Chain Mutation. Circulation 2019; 140:765-778. [PMID: 31315475 DOI: 10.1161/circulationaha.118.036965] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Restrictive cardiomyopathy is a rare heart disease associated with mutations in sarcomeric genes and with phenotypic overlap with hypertrophic cardiomyopathy. There is no approved therapy directed at the underlying cause. Here, we explore the potential of an interfering RNA (RNAi) therapeutic for a human sarcomeric mutation in MYL2 causative of restrictive cardiomyopathy in a mouse model. METHODS A short hairpin RNA (M7.8L) was selected from a pool for specificity and efficacy. Two groups of myosin regulatory light chain N47K transgenic mice were injected with M7.8L packaged in adeno-associated virus 9 at 3 days of age and 60 days of age. Mice were subjected to treadmill exercise and echocardiography after treatment to determine maximal oxygen uptake and left ventricular mass. At the end of treatment, heart, lung, liver, and kidney tissue was harvested to determine viral tropism and for transcriptomic and proteomic analysis. Cardiomyocytes were isolated for single-cell studies. RESULTS A one-time injection of AAV9-M7.8L RNAi in 3-day-old humanized regulatory light chain mutant transgenic mice silenced the mutated allele (RLC-47K) with minimal effects on the normal allele (RLC-47N) assayed at 16 weeks postinjection. AAV9-M7.8L RNAi suppressed the expression of hypertrophic biomarkers, reduced heart weight, and attenuated a pathological increase in left ventricular mass. Single adult cardiac myocytes from mice treated with AAV9-M7.8L showed partial restoration of contraction, relaxation, and calcium kinetics. In addition, cardiac stress protein biomarkers, such as calmodulin-dependent protein kinase II and the transcription activator Brg1 were reduced, suggesting recovery toward a healthy myocardium. Transcriptome analyses further revealed no significant changes of argonaute (AGO1, AGO2) and endoribonuclease dicer (DICER1) transcripts, and endogenous microRNAs were preserved, suggesting that the RNAi pathway was not saturated. CONCLUSIONS Our results show the feasibility, efficacy, and safety of RNAi therapeutics directed towards human restrictive cardiomyopathy. This is a promising step toward targeted therapy for a prevalent human disease.
Collapse
Affiliation(s)
- Kathia Zaleta-Rivera
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Alexandra Dainis
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | | | - Pablo Cordero
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Gabriel Rubio
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Ching Shang
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Jing Liu
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Thomas Finsterbach
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Victoria N Parikh
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Shirley Sutton
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Kinya Seo
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Nikita Sinha
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Nikhil Jain
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Yong Huang
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Roger J Hajjar
- Cardiovascular Institute, Cardiovascular Research Center at Icahn School of Medicine at Mount Sinai, New York, NY (R.J.H.)
| | - Mark A Kay
- Department of Genetics (M.A.K., E.A.A.), Stanford University School of Medicine, CA
- Department of Pediatrics (M.A.K.), Stanford University School of Medicine, CA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, FL (D.S.-C.)
| | - Beth L Pruitt
- Department of Mechanical Engineering, Stanford University, CA (A.J.S.R., B.L.P.)
| | - Matthew T Wheeler
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Euan A Ashley
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
- Department of Genetics (M.A.K., E.A.A.), Stanford University School of Medicine, CA
| |
Collapse
|
33
|
Multiparametric slice culture platform for the investigation of human cardiac tissue physiology. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 144:139-150. [DOI: 10.1016/j.pbiomolbio.2018.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/14/2018] [Accepted: 06/03/2018] [Indexed: 12/23/2022]
|
34
|
Affiliation(s)
- Evangelia G Kranias
- From the Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, OH (E.G.K.); and Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.H.).
| | - Roger J Hajjar
- From the Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, OH (E.G.K.); and Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.H.)
| |
Collapse
|
35
|
Fan J, Li H, Nie X, Yin Z, Zhao Y, Zhang X, Yuan S, Li Y, Chen C, Wang DW. MiR-665 aggravates heart failure via suppressing CD34-mediated coronary microvessel angiogenesis. Aging (Albany NY) 2018; 10:2459-2479. [PMID: 30243022 PMCID: PMC6188485 DOI: 10.18632/aging.101562] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/13/2018] [Indexed: 05/04/2023]
Abstract
BACKGROUND Heart failure (HF) is a major public health problem worldwide. The development of HF was related to coronary microvessel dysfunction. Whether miRNAs participate in HF by regulating coronary microvessel function remain unclear. METHODS The potential targets of miR-665 were predicted by rnahybrid software, then verified through anti-Ago2 co-immunoprecipitation, Western blotting and luciferase reporter assays. rAAV9 system was used to manipulate the expression of miR-665 in vivo. RESULTS Significant increase of miR-665 was observed in endothelial cells of human heart with heart failure. In vitro over-expression of miR-665 in endothelial cells resulted in decreased proliferation but enhanced apoptosis. rAAV-mediated delivery of miR-665 reduced coronary microvessel angiogenesis and cardiac microvessel density, then further impaired cardiac function in vivo. Furthermore, CD34 was confirmed as one of the miR-665 targets. Consistently, re-expression of CD34 attenuated miR-665-mediated damage effects in vitro and in vivo. We also found that Sp1 regulated miR-665 expression in endothelial cells. CONCLUSION Our findings demonstrated that miR-665 played an important role in heart failure via damaging coronary microvessel angiogenesis, and suggested that miRNA-based therapeutics may protect against coronary microvessel dysfunction and heart failure.
Collapse
Affiliation(s)
- Jiahui Fan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Huaping Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Xiang Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Zhongwei Yin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Yanru Zhao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Xudong Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Shuai Yuan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Yuying Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| |
Collapse
|
36
|
Poller W, Dimmeler S, Heymans S, Zeller T, Haas J, Karakas M, Leistner DM, Jakob P, Nakagawa S, Blankenberg S, Engelhardt S, Thum T, Weber C, Meder B, Hajjar R, Landmesser U. Non-coding RNAs in cardiovascular diseases: diagnostic and therapeutic perspectives. Eur Heart J 2018; 39:2704-2716. [PMID: 28430919 PMCID: PMC6454570 DOI: 10.1093/eurheartj/ehx165] [Citation(s) in RCA: 304] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/14/2017] [Accepted: 03/15/2017] [Indexed: 02/06/2023] Open
Abstract
Recent research has demonstrated that the non-coding genome plays a key role in genetic programming and gene regulation during development as well as in health and cardiovascular disease. About 99% of the human genome do not encode proteins, but are transcriptionally active representing a broad spectrum of non-coding RNAs (ncRNAs) with important regulatory and structural functions. Non-coding RNAs have been identified as critical novel regulators of cardiovascular risk factors and cell functions and are thus important candidates to improve diagnostics and prognosis assessment. Beyond this, ncRNAs are rapidly emgerging as fundamentally novel therapeutics. On a first level, ncRNAs provide novel therapeutic targets some of which are entering assessment in clinical trials. On a second level, new therapeutic tools were developed from endogenous ncRNAs serving as blueprints. Particularly advanced is the development of RNA interference (RNAi) drugs which use recently discovered pathways of endogenous short interfering RNAs and are becoming versatile tools for efficient silencing of protein expression. Pioneering clinical studies include RNAi drugs targeting liver synthesis of PCSK9 resulting in highly significant lowering of LDL cholesterol or targeting liver transthyretin (TTR) synthesis for treatment of cardiac TTR amyloidosis. Further novel drugs mimicking actions of endogenous ncRNAs may arise from exploitation of molecular interactions not accessible to conventional pharmacology. We provide an update on recent developments and perspectives for diagnostic and therapeutic use of ncRNAs in cardiovascular diseases, including atherosclerosis/coronary disease, post-myocardial infarction remodelling, and heart failure.
Collapse
Affiliation(s)
- Wolfgang Poller
- Department of Cardiology, CBF, CC11, Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Charite Centrum 11 (Cardiovascular Medicine), Hindenburgdamm 20, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Johann Wolfgang Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main, Germany
- DZHK, Site Rhein-Main, Frankfurt, Germany
| | - Stephane Heymans
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Tanja Zeller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Martinistrasse 52, Hamburg, Germany
- DZHK, Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Jan Haas
- Institute for Cardiomyopathies Heidelberg (ICH), Universitätsklinikum Heidelberg, Im Neuenheimer Feld 669, Heidelberg, Germany
- DZHK, Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Mahir Karakas
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Martinistrasse 52, Hamburg, Germany
- DZHK, Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - David-Manuel Leistner
- Department of Cardiology, CBF, CC11, Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Charite Centrum 11 (Cardiovascular Medicine), Hindenburgdamm 20, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany
| | - Philipp Jakob
- Department of Cardiology, CBF, CC11, Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Charite Centrum 11 (Cardiovascular Medicine), Hindenburgdamm 20, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany
| | - Shinichi Nakagawa
- RNA Biology Laboratory, RIKEN Advanced Research Institute, Wako, Saitama, Japan
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12-jo Nishi 6-chome, Kita-ku, Sapporo, Japan
| | - Stefan Blankenberg
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Martinistrasse 52, Hamburg, Germany
- DZHK, Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Stefan Engelhardt
- Institute for Pharmacology and Toxikology, Technische Universität München, Biedersteiner Strasse 29, München, Germany
- DZHK, Site Munich, Munich, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Christian Weber
- DZHK, Site Munich, Munich, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Pettenkoferstrasse 8a/9, Munich, Germany
| | - Benjamin Meder
- Institute for Cardiomyopathies Heidelberg (ICH), Universitätsklinikum Heidelberg, Im Neuenheimer Feld 669, Heidelberg, Germany
- DZHK, Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Roger Hajjar
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ulf Landmesser
- Department of Cardiology, CBF, CC11, Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Charite Centrum 11 (Cardiovascular Medicine), Hindenburgdamm 20, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany
- Berlin Institute of Health, Kapelle-Ufer 2, Berlin, Germany
| |
Collapse
|
37
|
Lahey R, Carley AN, Wang X, Glass CE, Accola KD, Silvestry S, O'Donnell JM, Lewandowski ED. Enhanced Redox State and Efficiency of Glucose Oxidation With miR Based Suppression of Maladaptive NADPH-Dependent Malic Enzyme 1 Expression in Hypertrophied Hearts. Circ Res 2018; 122:836-845. [PMID: 29386187 DOI: 10.1161/circresaha.118.312660] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 01/06/2023]
Abstract
RATIONALE Metabolic remodeling in hypertrophic hearts includes inefficient glucose oxidation via increased anaplerosis fueled by pyruvate carboxylation. Pyruvate carboxylation to malate through elevated ME1 (malic enzyme 1) consumes NADPH necessary for reduction of glutathione and maintenance of intracellular redox state. OBJECTIVE To elucidate upregulated ME1 as a potential maladaptive mechanism for inefficient glucose oxidation and compromised redox state in hypertrophied hearts. METHODS AND RESULTS ME1 expression was selectively inhibited, in vivo, via non-native miR-ME1 (miRNA specific to ME1) in pressure-overloaded rat hearts. Rats subjected to transverse aortic constriction (TAC) or Sham surgery received either miR-ME1 or PBS. Effects of ME1 suppression on anaplerosis and reduced glutathione (GSH) content were studied in isolated hearts supplied 13C-enriched substrate: palmitate, glucose, and lactate. Human myocardium collected from failing and nonfailing hearts during surgery enabled RT-qPCR confirmation of elevated ME1 gene expression in clinical heart failure versus nonfailing human hearts (P<0.04). TAC induced elevated ME1 content, but ME1 was lowered in hearts infused with miR-ME1 versus PBS. Although Sham miR-ME1 hearts showed no further reduction of inherently low anaplerosis in normal heart, miR-ME1 reduced anaplerosis in TAC to baseline: TAC miR-ME1=0.034±0.004; TAC PBS=0.081±0.005 (P<0.01). Countering elevated anaplerosis in TAC shifted pyruvate toward oxidation in the tricarboxylic acid cycle. Importantly, via the link to NADPH consumption by pyruvate carboxylation, ME1 suppression in TAC restored GSH content, reduced lactate production, and ultimately improved contractility. CONCLUSIONS A maladaptive increase in anaplerosis via ME1 in TAC is associated with reduced GSH content. Suppressing increased ME1 expression in hypertrophied rat hearts, which is also elevated in failing human hearts, reduced pyruvate carboxylation thereby normalizing anaplerosis, restoring GSH content, and reducing lactate accumulation. Reducing ME1 induced favorable metabolic shifts for carbohydrate oxidation, improving intracellular redox state and enhanced cardiac performance in pathological hypertrophy.
Collapse
Affiliation(s)
- Ryan Lahey
- From the Department of Internal Medicine and Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (A.N.C., E.D.L.); Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL (A.N.C., C.E.G., E.D.L.); Center for Cardiovascular Research, University of Illinois College of Medicine at Chicago (R.L., A.N.C., X.W., J.M.O., E.D.L.); and Translational Research Institute for Diabetes and Metabolism (C.E.G., E.D.L.) and Department of Surgery, Florida Hospital Cardiovascular Institute, Florida Hospital Transplant Center (K.D.A., S.S.), Florida Hospital, Orlando
| | - Andrew N Carley
- From the Department of Internal Medicine and Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (A.N.C., E.D.L.); Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL (A.N.C., C.E.G., E.D.L.); Center for Cardiovascular Research, University of Illinois College of Medicine at Chicago (R.L., A.N.C., X.W., J.M.O., E.D.L.); and Translational Research Institute for Diabetes and Metabolism (C.E.G., E.D.L.) and Department of Surgery, Florida Hospital Cardiovascular Institute, Florida Hospital Transplant Center (K.D.A., S.S.), Florida Hospital, Orlando
| | - Xuerong Wang
- From the Department of Internal Medicine and Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (A.N.C., E.D.L.); Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL (A.N.C., C.E.G., E.D.L.); Center for Cardiovascular Research, University of Illinois College of Medicine at Chicago (R.L., A.N.C., X.W., J.M.O., E.D.L.); and Translational Research Institute for Diabetes and Metabolism (C.E.G., E.D.L.) and Department of Surgery, Florida Hospital Cardiovascular Institute, Florida Hospital Transplant Center (K.D.A., S.S.), Florida Hospital, Orlando
| | - Carley E Glass
- From the Department of Internal Medicine and Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (A.N.C., E.D.L.); Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL (A.N.C., C.E.G., E.D.L.); Center for Cardiovascular Research, University of Illinois College of Medicine at Chicago (R.L., A.N.C., X.W., J.M.O., E.D.L.); and Translational Research Institute for Diabetes and Metabolism (C.E.G., E.D.L.) and Department of Surgery, Florida Hospital Cardiovascular Institute, Florida Hospital Transplant Center (K.D.A., S.S.), Florida Hospital, Orlando
| | - Kevin D Accola
- From the Department of Internal Medicine and Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (A.N.C., E.D.L.); Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL (A.N.C., C.E.G., E.D.L.); Center for Cardiovascular Research, University of Illinois College of Medicine at Chicago (R.L., A.N.C., X.W., J.M.O., E.D.L.); and Translational Research Institute for Diabetes and Metabolism (C.E.G., E.D.L.) and Department of Surgery, Florida Hospital Cardiovascular Institute, Florida Hospital Transplant Center (K.D.A., S.S.), Florida Hospital, Orlando
| | - Scott Silvestry
- From the Department of Internal Medicine and Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (A.N.C., E.D.L.); Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL (A.N.C., C.E.G., E.D.L.); Center for Cardiovascular Research, University of Illinois College of Medicine at Chicago (R.L., A.N.C., X.W., J.M.O., E.D.L.); and Translational Research Institute for Diabetes and Metabolism (C.E.G., E.D.L.) and Department of Surgery, Florida Hospital Cardiovascular Institute, Florida Hospital Transplant Center (K.D.A., S.S.), Florida Hospital, Orlando
| | - J Michael O'Donnell
- From the Department of Internal Medicine and Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (A.N.C., E.D.L.); Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL (A.N.C., C.E.G., E.D.L.); Center for Cardiovascular Research, University of Illinois College of Medicine at Chicago (R.L., A.N.C., X.W., J.M.O., E.D.L.); and Translational Research Institute for Diabetes and Metabolism (C.E.G., E.D.L.) and Department of Surgery, Florida Hospital Cardiovascular Institute, Florida Hospital Transplant Center (K.D.A., S.S.), Florida Hospital, Orlando
| | - E Douglas Lewandowski
- From the Department of Internal Medicine and Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (A.N.C., E.D.L.); Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL (A.N.C., C.E.G., E.D.L.); Center for Cardiovascular Research, University of Illinois College of Medicine at Chicago (R.L., A.N.C., X.W., J.M.O., E.D.L.); and Translational Research Institute for Diabetes and Metabolism (C.E.G., E.D.L.) and Department of Surgery, Florida Hospital Cardiovascular Institute, Florida Hospital Transplant Center (K.D.A., S.S.), Florida Hospital, Orlando.
| |
Collapse
|
38
|
Abstract
Silencing of cardiac genes by RNA interference (RNAi) has developed into a powerful new method to treat cardiac diseases. Small interfering (si)RNAs are the inducers of RNAi, but cultured primary cardiomyocytes and heart are highly resistant to siRNA transfection. This can be overcome by delivery of small hairpin (sh)RNAs or artificial microRNA (amiRNAs) by cardiotropic adeno-associated virus (AAV) vectors. Here we describe as example of the silencing of a cardiac gene, the generation and cloning of shRNA, and amiRNAs directed against the cardiac protein phospholamban. We further describe the generation of AAV shuttle plasmids with self complementary vector genomes, the production of AAV vectors in roller bottles, and their purification via iodixanol gradient centrifugation and concentration with filter systems. Finally we describe the preparation of primary neonatal rat cardiomyocytes (PNRC), the transduction of PNRC with AAV vectors, and the maintenance of the transduced cell culture.
Collapse
Affiliation(s)
- Henry Fechner
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355, Berlin, Germany.
| | - Roland Vetter
- Institute of Clinical Pharmacology & Toxicology, Charité - Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117, Berlin, Germany
| | - Jens Kurreck
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Wolfgang Poller
- Department of Cardiology & Pneumology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany
| |
Collapse
|
39
|
Kaneko M, Yamamoto H, Sakai H, Kamada Y, Tanaka T, Fujiwara S, Yamamoto S, Takahagi H, Igawa H, Kasai S, Noda M, Inui M, Nishimoto T. A pyridone derivative activates SERCA2a by attenuating the inhibitory effect of phospholamban. Eur J Pharmacol 2017; 814:1-8. [DOI: 10.1016/j.ejphar.2017.07.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 01/26/2023]
|
40
|
Voit A, Patel V, Pachon R, Shah V, Bakhutma M, Kohlbrenner E, McArdle JJ, Dell'Italia LJ, Mendell JR, Xie LH, Hajjar RJ, Duan D, Fraidenraich D, Babu GJ. Reducing sarcolipin expression mitigates Duchenne muscular dystrophy and associated cardiomyopathy in mice. Nat Commun 2017; 8:1068. [PMID: 29051551 PMCID: PMC5648780 DOI: 10.1038/s41467-017-01146-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 08/22/2017] [Indexed: 01/16/2023] Open
Abstract
Sarcolipin (SLN) is an inhibitor of the sarco/endoplasmic reticulum (SR) Ca2+ ATPase (SERCA) and is abnormally elevated in the muscle of Duchenne muscular dystrophy (DMD) patients and animal models. Here we show that reducing SLN levels ameliorates dystrophic pathology in the severe dystrophin/utrophin double mutant (mdx:utr -/-) mouse model of DMD. Germline inactivation of one allele of the SLN gene normalizes SLN expression, restores SERCA function, mitigates skeletal muscle and cardiac pathology, improves muscle regeneration, and extends the lifespan. To translate our findings into a therapeutic strategy, we knock down SLN expression in 1-month old mdx:utr -/- mice via adeno-associated virus (AAV) 9-mediated RNA interference. The AAV treatment markedly reduces SLN expression, attenuates muscle pathology and improves diaphragm, skeletal muscle and cardiac function. Taken together, our findings suggest that SLN reduction is a promising therapeutic approach for DMD.
Collapse
Affiliation(s)
- Antanina Voit
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Vishwendra Patel
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Ronald Pachon
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Vikas Shah
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Mohammad Bakhutma
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Erik Kohlbrenner
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Joseph J McArdle
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Louis J Dell'Italia
- Department of Medicine, University of Alabama at Birmingham, and Birmingham VA Medical Center, Birmingham, AL, 35294, USA
| | - Jerry R Mendell
- Department of Pediatrics and Department of Neurology, Ohio State University Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, Neurology, Bioengineering, Biomedical Sciences, The University of Missouri, Columbia, MO, 65212, USA
| | - Diego Fraidenraich
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Gopal J Babu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA.
| |
Collapse
|
41
|
Bera A, Sen D. Promise of adeno-associated virus as a gene therapy vector for cardiovascular diseases. Heart Fail Rev 2017; 22:795-823. [DOI: 10.1007/s10741-017-9622-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
42
|
Meyer K, Hodwin B, Ramanujam D, Engelhardt S, Sarikas A. Essential Role for Premature Senescence of Myofibroblasts in Myocardial Fibrosis. J Am Coll Cardiol 2017; 67:2018-28. [PMID: 27126529 DOI: 10.1016/j.jacc.2016.02.047] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 02/08/2016] [Accepted: 02/15/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Fibrosis is a hallmark of many myocardial pathologies and contributes to distorted organ architecture and function. Recent studies have identified premature senescence as a regulatory mechanism of tissue fibrosis, but its relevance in the heart remains to be established. OBJECTIVES This study investigated the role of premature senescence in myocardial fibrosis. METHODS Murine models of cardiac diseases and human heart biopsies were analyzed for characteristics of premature senescence and fibrosis. Loss-of-function and gain-of-function models of premature senescence were used to determine its pathophysiological role in myocardial fibrosis. RESULTS Senescence markers p21(CIP1/WAF1), senescence-associated ß-galactosidase (SA-ß-gal), and p16(INK4a) were increased 2-, 8-, and 20-fold (n = 5 to 7; p < 0.01), respectively, in perivascular fibrotic areas after transverse aortic constriction compared with sham-treated control subjects. Similar results were observed with cardiomyocyte-specific β1-adrenoceptor transgenic mice and human heart biopsies. Senescent cells were positive for platelet-derived growth factor receptor-α, vimentin, and α-smooth muscle actin, specifying myofibroblasts as the predominant cell population undergoing premature senescence in the heart. Inactivation of the premature senescence program by genetic ablation of p53 and p16(INK4a) (Trp53(-/-)Cdkn2a(-/-) mice) resulted in aggravated fibrosis after transverse aortic constriction, when compared with wild-type control subjects (49 ± 4.9% vs. 33 ± 2.7%; p < 0.01), and was associated with impaired cardiac function. Conversely, cardiac-specific expression of CCN1 (CYR61), a potent inducer of premature senescence, by adeno-associated virus serotype 9 gene transfer, resulted in ∼50% reduction of perivascular fibrosis after transverse aortic constriction, when compared with mock- or dominant-negative CCN1-infected control subjects, and improved cardiac function. CONCLUSIONS Our data establish premature senescence of myofibroblasts as an essential antifibrotic mechanism and potential therapeutic target in myocardial fibrosis.
Collapse
Affiliation(s)
- Kathleen Meyer
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Bettina Hodwin
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
| | - Deepak Ramanujam
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Antonio Sarikas
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
43
|
Sánchez-Gómez MC, García-Mejía KA, Pérez-Díaz Conti M, Díaz-Rosas G, Palma-Lara I, Sánchez-Urbina R, Klünder-Klünder M, Botello-Flores JA, Balderrábano-Saucedo NA, Contreras-Ramos A. MicroRNAs Association in the Cardiac Hypertrophy Secondary to Complex Congenital Heart Disease in Children. Pediatr Cardiol 2017; 38:991-1003. [PMID: 28382463 DOI: 10.1007/s00246-017-1607-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 03/20/2017] [Indexed: 12/14/2022]
Abstract
Complex congenital heart disease (CHD) affects cardiac blood flow, generating a pressure overload in the compromised ventricles and provoking hypertrophy that over time will induce myocardial dysfunction and cause a potential risk of imminent death. Therefore, the early diagnosis of complex CHD is paramount during the first year of life, with surgical treatment of patients favoring survival. In the present study, we analyzed cardiac tissue and plasma of children with cardiac hypertrophy (CH) secondary to CHD for the expression of 11 miRNAs specific to CH in adults. The results were compared with the miRNA expression patterns in tissue and blood of healthy children. In this way, we determined that miRNAs 1, 18b, 21, 23b, 133a, 195, and 208b constitute the expression profile of the cardiac tissue of children with CHD. Meanwhile, miRNAs 21, 23a, 23b, and 24 can be considered specific biomarkers for the diagnosis of CH in infants with CHD. These results suggest that CH secondary to CHD in children differs in its mechanism from that described for adult hypertrophy, offering a new perspective to study the development of this pathology and to determine the potential of hypertrophic miRNAs to be biomarkers for early CH.
Collapse
Affiliation(s)
- Ma C Sánchez-Gómez
- Laboratory of Developmental Biology Research and Experimental Teratogenicity, Children's Hospital of Mexico Federico Gomez (HIMFG), CP 06720, Mexico City, Mexico.,School of Medicine, National Polytechnic Institute (IPN), Mexico City, Mexico
| | - K A García-Mejía
- Laboratory of Developmental Biology Research and Experimental Teratogenicity, Children's Hospital of Mexico Federico Gomez (HIMFG), CP 06720, Mexico City, Mexico
| | | | - G Díaz-Rosas
- Laboratory of Developmental Biology Research and Experimental Teratogenicity, Children's Hospital of Mexico Federico Gomez (HIMFG), CP 06720, Mexico City, Mexico
| | - I Palma-Lara
- School of Medicine, National Polytechnic Institute (IPN), Mexico City, Mexico
| | - R Sánchez-Urbina
- Laboratory of Developmental Biology Research and Experimental Teratogenicity, Children's Hospital of Mexico Federico Gomez (HIMFG), CP 06720, Mexico City, Mexico
| | | | - J A Botello-Flores
- Laboratory of Developmental Biology Research and Experimental Teratogenicity, Children's Hospital of Mexico Federico Gomez (HIMFG), CP 06720, Mexico City, Mexico
| | | | - A Contreras-Ramos
- Laboratory of Developmental Biology Research and Experimental Teratogenicity, Children's Hospital of Mexico Federico Gomez (HIMFG), CP 06720, Mexico City, Mexico.
| |
Collapse
|
44
|
Paul P, Chakraborty A, Sarkar D, Langthasa M, Rahman M, Bari M, Singha RS, Malakar AK, Chakraborty S. Interplay between miRNAs and human diseases. J Cell Physiol 2017; 233:2007-2018. [PMID: 28181241 DOI: 10.1002/jcp.25854] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs) are endogenous, non-coding RNAs, which have evoked a great deal of interest due to their importance in many aspects of homeostasis and diseases. MicroRNAs are stable and are essential components of gene regulatory networks. They play a crucial role in healthy individuals and their dysregulations have also been implicated in a wide range of diseases, including diabetes, cardiovascular disease, kidney disease, and cancer. This review summarized the current understanding of interactions between miRNAs and different diseases and their role in disease diagnosis and therapy.
Collapse
Affiliation(s)
- Prosenjit Paul
- Department of Biotechnology, Assam University, Silchar, Assam, India
| | | | - Debasree Sarkar
- Department of Biotechnology, Assam University, Silchar, Assam, India
| | | | - Musfhia Rahman
- Department of Biotechnology, Assam University, Silchar, Assam, India
| | - Minakshi Bari
- Department of Biotechnology, Assam University, Silchar, Assam, India
| | | | | | | |
Collapse
|
45
|
Zhou L, Li R, Liu C, Sun T, Htet Aung LH, Chen C, Gao J, Zhao Y, Wang K. Foxo3a inhibits mitochondrial fission and protects against doxorubicin-induced cardiotoxicity by suppressing MIEF2. Free Radic Biol Med 2017; 104:360-370. [PMID: 28137654 DOI: 10.1016/j.freeradbiomed.2017.01.037] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/24/2017] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
Abstract
Doxorubicin (DOX) as a chemotherapeutic drug is widely used to treat a variety of human tumors. However, a major factor limiting its clinical use is its cardiotoxicity. The molecular components and detailed mechanisms regulating DOX-induced cardiotoxicity remain largely unidentified. Here we report that Foxo3a is downregulated in the cardiomyocyte and mouse heart in response to DOX treatment. Foxo3a attenuates DOX-induced mitochondrial fission and apoptosis in cardiomyocytes. Cardiac specific Foxo3a transgenic mice show reduced mitochondrial fission, apoptosis and cardiotoxicity upon DOX administration. Furthermore, Foxo3a directly targets mitochondrial dynamics protein of 49kDa (MIEF2) and suppresses its expression at transcriptional level. Knockdown of MIEF2 reduces DOX-induced mitochondrial fission and apoptosis in cardiomyocytes and in vivo. Also, knockdown of MIEF2 protects heart from DOX-induced cardiotoxicity. Our study identifies a novel pathway composed of Foxo3a and MIEF2 that mediates DOX cardiotoxicity. This discovery provides a promising therapeutic strategy for the treatment of cancer therapy and cardioprotection.
Collapse
Affiliation(s)
- Luyu Zhou
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Ruibei Li
- School of Professional Studies, Northwestern University, Chicago, IL 60611, USA
| | - Cuiyun Liu
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Teng Sun
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Lynn Htet Htet Aung
- College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Chao Chen
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Jinning Gao
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yanfang Zhao
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Kun Wang
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
46
|
Yin Z, Zhao Y, Li H, Yan M, Zhou L, Chen C, Wang DW. miR-320a mediates doxorubicin-induced cardiotoxicity by targeting VEGF signal pathway. Aging (Albany NY) 2016; 8:192-207. [PMID: 26837315 PMCID: PMC4761722 DOI: 10.18632/aging.100876] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Vascular homeostasis abnormalities may involve in doxorubicin induced cardiotoxicity. Methods Enhanced cardiac miR-320a expression, reduced cardiac microvessel density and impaired cardiac function were observed in mice treated by anthracycline doxorubicin. To further explore the role of miR-320a in doxorubicin induced cardiotoxicity, microRNA mimics/inhibitor in vitro and rAAV administration in vivo were employed in mice. Results Knockdown of miR-320a not only resulted in enhanced proliferation and inhibited apoptosis in cultured endothelial cells, but also attenuated cardiac abnormalities induced by doxorubicin. On the contrary, overexpression of miR-320a enhanced apoptosis in vitro, and aggravated vessel abnormalities in heart and subsequent cardiac dysfunction in mice. Furthermore, Western blot assays showed that VEGF-A was a potential target of miR-320a, which was verified by anti-Ago2 co-immunoprecipitation. Moreover, as same as miR-320a, siRNA against VEGF-A reinforced doxorubicin induced endothelial cells injury. Finally, the negative effects of miR-320a on vascular homeostasis and cardiac function were alleviated by VEGF-A re-expression in doxorubicin treated mice. Conclusion Our observations demonstrate that miR-320a play important roles in doxorubicin induced cardiotoxicity via vessel homeostasis in heart and thus, inhibition of miR-320a may be applied to the treatment of cardiac dysfunction induced by anthracycline.
Collapse
Affiliation(s)
- Zhongwei Yin
- Division of Cardiology, Departments of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yanru Zhao
- Division of Cardiology, Departments of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Huaping Li
- Division of Cardiology, Departments of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Mengwen Yan
- Division of Cardiology, Departments of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Ling Zhou
- Division of Cardiology, Departments of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Chen Chen
- Division of Cardiology, Departments of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Dao Wen Wang
- Division of Cardiology, Departments of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| |
Collapse
|
47
|
Wang H, Cai J. The role of microRNAs in heart failure. Biochim Biophys Acta Mol Basis Dis 2016; 1863:2019-2030. [PMID: 27916680 DOI: 10.1016/j.bbadis.2016.11.034] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/26/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
MicroRNAs are small non-coding RNA molecules that regulate gene expression by inhibiting mRNA translation and/or inducing mRNA degradation. In the past decade, many in vitro and in vivo studies have explored the involvement of microRNAs in various cardiovascular diseases. In this paper, studies focused upon the target genes and functionality of miRNAs in the pathophysiological processes of heart failure are reviewed. The selected miRNAs are categorized according to the biological relevance of their target genes in relation to four cardiovascular pathologies, namely angiogenesis, cardiac hypertrophy, fibrosis and apoptosis. This review illustrates the involvement of miRNAs in different biological signaling pathways and provides an overview of current understanding of the roles of miRNAs in cardiovascular health and diseases. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.
Collapse
Affiliation(s)
- Hongjiang Wang
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China.
| | - Jun Cai
- State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Diseases of China, Chinese Academy of Medical Sciences and Peking Union Medical College, Hypertension Center, Fuwai Hospital, Xicheng District, North Lishi Road No. 167, Beijing 100037, China.
| |
Collapse
|
48
|
Duan Q, Ni L, Wang P, Chen C, Yang L, Ma B, Gong W, Cai Z, Zou M, Wang DW. Deregulation of XBP1 expression contributes to myocardial vascular endothelial growth factor-A expression and angiogenesis during cardiac hypertrophy in vivo. Aging Cell 2016; 15:625-33. [PMID: 27133203 PMCID: PMC4933664 DOI: 10.1111/acel.12460] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2016] [Indexed: 01/18/2023] Open
Abstract
Endoplasmic reticulum (ER) stress has been reported to be involved in many cardiovascular diseases such as atherosclerosis, diabetes, myocardial ischemia, and hypertension that ultimately result in heart failure. XBP1 is a key ER stress signal transducer and an important pro‐survival factor of the unfolded protein response (UPR) in mammalian cells. The aim of this study was to establish a role for XBP1 in the deregulation of pro‐angiogenic factor VEGF expression and potential regulatory mechanisms in hypertrophic and failing heart. Western blots showed that myocardial XBP1s protein was significantly increased in both isoproterenol (ISO)‐induced and pressure‐overload‐induced hypertrophic and failing heart compared to normal control. Furthermore, XBP1 silencing exacerbates ISO‐induced cardiac dysfunction along with a reduction of myocardial capillary density and cardiac expression of pro‐angiogenic factor VEGF‐A in vivo. Consistently, experiments in cultured cardiomyocytes H9c2 (2‐1) cells showed that UPR‐induced VEGF‐A upregulation was determined by XBP1 expression level. Importantly, VEGF‐A expression was increased in failing human heart tissue and blood samples and was correlated with the levels of XBP1. These results suggest that XBP1 regulates VEGF‐mediated cardiac angiogenesis, which contributes to the progression of adaptive hypertrophy, and might provide novel targets for prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Quanlu Duan
- Division of Cardiology, Department Internal Medicine, Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 People's Republic of China
| | - Li Ni
- Division of Cardiology, Department Internal Medicine, Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 People's Republic of China
| | - Peihua Wang
- Division of Cardiology, Department Internal Medicine, Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 People's Republic of China
| | - Chen Chen
- Division of Cardiology, Department Internal Medicine, Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 People's Republic of China
| | - Lei Yang
- Division of Cardiology, Department Internal Medicine, Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 People's Republic of China
| | - Ben Ma
- Division of Cardiology, Department Internal Medicine, Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 People's Republic of China
| | - Wei Gong
- Division of Cardiology, Department Internal Medicine, Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 People's Republic of China
| | - Zhejun Cai
- Division of Cardiology, Department Internal Medicine, Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 People's Republic of China
| | - Ming‐Hui Zou
- Center for Molecular and Translational Medicine Georgia State University Atlanta 30303 GA USA
| | - Dao Wen Wang
- Division of Cardiology, Department Internal Medicine, Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 People's Republic of China
| |
Collapse
|
49
|
Kopechek JA, Carson AR, McTiernan CF, Chen X, Klein EC, Villanueva FS. Cardiac Gene Expression Knockdown Using Small Inhibitory RNA-Loaded Microbubbles and Ultrasound. PLoS One 2016; 11:e0159751. [PMID: 27471848 PMCID: PMC4966949 DOI: 10.1371/journal.pone.0159751] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 07/07/2016] [Indexed: 01/29/2023] Open
Abstract
RNA interference has potential therapeutic value for cardiac disease, but targeted delivery of interfering RNA is a challenge. Custom designed microbubbles, in conjunction with ultrasound, can deliver small inhibitory RNA to target tissues in vivo. The efficacy of cardiac RNA interference using a microbubble-ultrasound theranostic platform has not been demonstrated in vivo. Therefore, our objective was to test the hypothesis that custom designed microbubbles and ultrasound can mediate effective delivery of small inhibitory RNA to the heart. Microbubble and ultrasound mediated cardiac RNA interference was tested in transgenic mice displaying cardiac-restricted luciferase expression. Luciferase expression was assayed in select tissues of untreated mice (n = 14). Mice received intravenous infusion of cationic microbubbles bearing small inhibitory RNA directed against luciferase (n = 9) or control RNA (n = 8) during intermittent cardiac-directed ultrasound at mechanical index of 1.6. Simultaneous echocardiography in a separate group of mice (n = 3) confirmed microbubble destruction and replenishment during treatment. Three days post treatment, cardiac luciferase messenger RNA and protein levels were significantly lower in ultrasound-treated mice receiving microbubbles loaded with small inhibitory RNA directed against luciferase compared to mice receiving microbubbles bearing control RNA (23±7% and 33±7% of control mice, p<0.01 and p = 0.03, respectively). Passive cavitation detection focused on the heart confirmed that insonification resulted in inertial cavitation. In conclusion, small inhibitory RNA-loaded microbubbles and ultrasound directed at the heart significantly reduced the expression of a reporter gene. Ultrasound-targeted destruction of RNA-loaded microbubbles may be an effective image-guided strategy for therapeutic RNA interference in cardiac disease.
Collapse
Affiliation(s)
- Jonathan A. Kopechek
- Dept. of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Dept. of Bioengineering, University of Louisville, Louisville, KY, United States of America
| | - Andrew R. Carson
- Dept. of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Charles F. McTiernan
- Dept. of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Xucai Chen
- Dept. of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Edwin C. Klein
- Dept. of Laboratory Animal Resources, University of Pittsburgh, Pittsburgh, PA, United States of America
| | | |
Collapse
|
50
|
Lother A, Hein L. Pharmacology of heart failure: From basic science to novel therapies. Pharmacol Ther 2016; 166:136-49. [PMID: 27456554 DOI: 10.1016/j.pharmthera.2016.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/08/2016] [Indexed: 01/10/2023]
Abstract
Chronic heart failure is one of the leading causes for hospitalization in the United States and Europe, and is accompanied by high mortality. Current pharmacological therapy of chronic heart failure with reduced ejection fraction is largely based on compounds that inhibit the detrimental action of the adrenergic and the renin-angiotensin-aldosterone systems on the heart. More than one decade after spironolactone, two novel therapeutic principles have been added to the very recently released guidelines on heart failure therapy: the HCN-channel inhibitor ivabradine and the combined angiotensin and neprilysin inhibitor valsartan/sacubitril. New compounds that are in phase II or III clinical evaluation include novel non-steroidal mineralocorticoid receptor antagonists, guanylate cyclase activators or myosine activators. A variety of novel candidate targets have been identified and the availability of gene transfer has just begun to accelerate translation from basic science to clinical application. This review provides an overview of current pharmacology and pharmacotherapy in chronic heart failure at three stages: the updated clinical guidelines of the American Heart Association and the European Society of Cardiology, new drugs which are in clinical development, and finally innovative drug targets and their mechanisms in heart failure which are emerging from preclinical studies will be discussed.
Collapse
Affiliation(s)
- Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Heart Center, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|