1
|
Coughlin TM, Makarewich CA. Emerging roles for microproteins as critical regulators of endoplasmic reticulum function and cellular homeostasis. Semin Cell Dev Biol 2025; 170:103608. [PMID: 40245464 PMCID: PMC12065929 DOI: 10.1016/j.semcdb.2025.103608] [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: 11/19/2024] [Revised: 02/20/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025]
Abstract
The endoplasmic reticulum (ER) is a multifunctional organelle essential for key cellular processes including protein synthesis, calcium homeostasis, and the cellular stress response. It is composed of distinct domains, such as the rough and smooth ER, as well as membrane regions that facilitate direct communication with other organelles, enabling its diverse functions. While many well-characterized ER proteins contribute to these processes, recent studies have revealed a previously underappreciated class of small proteins that play critical regulatory roles. Microproteins, typically under 100 amino acids in length, were historically overlooked due to size-based biases in genome annotation and often misannotated as noncoding RNAs. Advances in ribosome profiling, mass spectrometry, and computational approaches have now enabled the discovery of numerous previously unrecognized microproteins, significantly expanding our understanding of the proteome. While some ER-associated microproteins, such as phospholamban and sarcolipin, were identified decades ago, newly discovered microproteins share similar fundamental characteristics, underscoring the need to refine our understanding of the coding potential of the genome. Molecular studies have demonstrated that ER microproteins play essential roles in calcium regulation, ER stress response, organelle communication, and protein translocation. Moreover, growing evidence suggests that ER microproteins contribute to cellular homeostasis and are implicated in disease processes, including cardiovascular disease and cancer. This review examines the shared and unique functions of ER microproteins, their implications for health and disease, and their potential as therapeutic targets for conditions associated with ER dysfunction.
Collapse
Affiliation(s)
- Taylor M Coughlin
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Catherine A Makarewich
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
2
|
Li L, Lu M, Guo L, Zhang X, Liu Q, Zhang M, Gao J, Xu M, Lu Y, Zhang F, Li Y, Zhang R, Liu X, Pan S, Zhang X, Li Z, Chen Y, Su X, Zhang N, Guo W, Yang T, Chen J, Qin Y, Zhang Z, Cui W, Yu L, Gu Y, Yang H, Xu X, Wang J, Burns CE, Burns CG, Han K, Zhao L, Fan G, Su Y. An organ-wide spatiotemporal transcriptomic and cellular atlas of the regenerating zebrafish heart. Nat Commun 2025; 16:3716. [PMID: 40253397 PMCID: PMC12009352 DOI: 10.1038/s41467-025-59070-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 04/10/2025] [Indexed: 04/21/2025] Open
Abstract
Adult zebrafish robustly regenerate injured hearts through a complex orchestration of molecular and cellular activities. However, this remarkable process, which is largely non-existent in humans, remains incompletely understood. Here, we utilize integrated spatial transcriptomics (Stereo-seq) and single-cell RNA-sequencing (scRNA-seq) to generate a spatially-resolved molecular and cellular atlas of regenerating zebrafish heart across eight stages. We characterize the cascade of cardiomyocyte cell states responsible for producing regenerated myocardium and explore a potential role for tpm4a in cardiomyocyte re-differentiation. Moreover, we uncover the activation of ifrd1 and atp6ap2 genes as a unique feature of regenerative hearts. Lastly, we reconstruct a 4D "virtual regenerating heart" comprising 569,896 cells/spots derived from 36 scRNA-seq libraries and 224 Stereo-seq slices. Our comprehensive atlas serves as a valuable resource to the cardiovascular and regeneration scientific communities and their ongoing efforts to understand the molecular and cellular mechanisms underlying vertebrate heart regeneration.
Collapse
Affiliation(s)
- Lei Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
| | - Meina Lu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Lidong Guo
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuejiao Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Qun Liu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Meiling Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Junying Gao
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Mengyang Xu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
| | - Yijian Lu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Fang Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Yao Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Ruihua Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xiawei Liu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Shanshan Pan
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xianghui Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Zhen Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Yadong Chen
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xiaoshan Su
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Nannan Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Wenjie Guo
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Tao Yang
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | - Jing Chen
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | - Yating Qin
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | | | - Wei Cui
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Lindong Yu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Ying Gu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Huanming Yang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Xun Xu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Caroline E Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - C Geoffrey Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Kai Han
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark.
| | - Long Zhao
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
- College of Fisheries, Ocean University of China, Qingdao, 266003, China.
| | - Guangyi Fan
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China.
- BGI Research, Sanya, 572025, China.
- BGI Research, Hangzhou, 310030, China.
| | - Ying Su
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
3
|
Deiman FE, de Brouwer R, Baumhove L, Bomer N, Grote Beverborg N, van der Meer P. Identification of disease-specific pathways and modifiers in phospholamban R14del cardiomyopathy: rationale, design and baseline characteristics of DECIPHER-PLN cohort. Neth Heart J 2025; 33:112-119. [PMID: 40048085 PMCID: PMC11953489 DOI: 10.1007/s12471-025-01941-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Phospholamban (PLN) p.Arg14del (R14del, R14∆/+) is the most commonly identified pathogenic variant that causes cardiomyopathy in the Netherlands. Many disease characteristics are still unclear, including the phenotypic triggers, disease progression and disease-specific biomarkers. We aim to gain a better understanding of the R14∆/+ pathophysiology by establishing a cohort across the R14∆/+ disease spectrum. METHODS The Disease spECifIc PatHways and modifiERs in PhosphoLambaN r14del cardiomyopathy (DECIPHER-PLN) cohort includes 101 participants, categorised as unaffected R14∆/+ (n = 21), early affected R14∆/+ (n = 42), end-stage R14∆/+ (n = 28) and heart failure (HF) of another aetiology (n = 10). R14∆/+ category was based on left ventricular ejection fraction, HF symptoms, electrocardiogram (ECG) and N‑terminal pro-brain natriuretic peptide concentrations. Of the 91 included R14∆/+ carriers, 46 (51%) were female, with a mean age of 55 years (standard deviation: 14). Low-voltage ECG older age, arrhythmias, and conduction and repolarisation abnormalities were common in (early) affected R14∆/+ carriers. Serum and plasma were collected from all participants. Induced pluripotent stem cells were generated from fibroblasts of end-stage R14∆/+ patients and unaffected R14∆/+ family members (n = 4) and differentiated into cardiomyocytes. Explanted heart tissue was obtained from R14∆/+ patients undergoing cardiac surgery and patients with other HF aetiologies as control. Abnormal PLN protein localisation was confirmed in R14∆/+ carriers. CONCLUSION DECIPHER-PLN comprises R14∆/+ carriers across the disease and non-disease spectrum and can be used to identify disease-specific biological pathways and modifiers that play a role in R14∆/+ cardiomyopathy. Using a multi-omics approach and in vitro disease modelling, we aim to identify novel biomarkers and improve our understanding of R14∆/+ pathophysiology. Material is available upon request.
Collapse
Affiliation(s)
- Frederik E Deiman
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Remco de Brouwer
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Lukas Baumhove
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Nils Bomer
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Niels Grote Beverborg
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands.
| |
Collapse
|
4
|
Hamstra SI, Geromella MS, Tiidus P, Klentrou P, MacPherson REK, Fajardo VA. Subtherapeutic lithium supplementation causes physiological eccentric cardiac hypertrophy in young-adult wild-type male mice. Physiol Rep 2025; 13:e70299. [PMID: 40170524 PMCID: PMC11962211 DOI: 10.14814/phy2.70299] [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] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/03/2025] Open
Abstract
Six weeks of low-dose lithium (Li) supplementation has been shown to improve the activity of cardiac sarco(endo)plasmic reticulum calcium (Ca2+)-ATPase (SERCA) in C57BL/6J wild-type (WT) male mice. Improvements in myocardial SERCA function can lead to improvements in systolic and diastolic function in various rodent models. In this study, we tested the hypothesis that 12 weeks of subtherapeutic Li supplementation (10 mg/kg/day) would enhance SERCA function and positively influence cardiac contractility and morphology. Cardiac function and morphology were assessed using high-frequency ultrasound in the final week of Li treatment. Subsequently, SERCA activity, Ca2+ uptake assays, and Western blotting for glycogen synthase kinase-3β, SERCA2, and its inhibitor phospholamban (PLN) were performed on isolated left ventricle tissue. After 12 weeks of subtherapeutic Li supplementation, the heart underwent eccentric remodeling, exhibited by increased left ventricle internal diameter and volumes during systole and diastole, ultimately leading to greater stroke volume. However, we did not find any specific alterations in systolic or diastolic functional measures; nor were there any changes in SERCA activity and its content relative to PLN after Li supplementation. Thus, while Li supplementation appears to positively influence cardiac morphology to increase stroke volume, these changes are independent of changes to SERCA function.
Collapse
Affiliation(s)
- Sophie I. Hamstra
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| | - Mia S. Geromella
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| | - Peter Tiidus
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
| | - Panagiota Klentrou
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| | | | - Val A. Fajardo
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| |
Collapse
|
5
|
Sorella A, Galanti K, Iezzi L, Gallina S, Mohammed SF, Sekhri N, Akhtar MM, Prasad SK, Chahal CAA, Ricci F, Khanji MY. Diagnosis and management of dilated cardiomyopathy: a systematic review of clinical practice guidelines and recommendations. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2025; 11:206-222. [PMID: 39674807 PMCID: PMC11879293 DOI: 10.1093/ehjqcco/qcae109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 12/16/2024]
Abstract
Dilated cardiomyopathy (DCM) is extensively discussed in numerous expert consensus documents and international guidelines, with differing recommendations. To support clinicians in daily practice and decision-making, we conducted a systematic review of key guidelines and recommendations concerning the diagnosis and clinical management of DCM. Our research encompassed MEDLINE and EMBASE databases for relevant articles published, as well as the websites of relevant scientific societies. We identified two guidelines and one scientific statement that met stringent criteria, thereby qualifying them for detailed systematic analysis. Our review revealed consensus on several key aspects: the definition of DCM, the use of B-type natriuretic peptides and high-sensitivity troponin in laboratory testing, the essential role of multimodality cardiovascular imaging for initial diagnosis, genetic counselling, and the management of advanced disease. Nonetheless, notable areas of variation included the formation of multidisciplinary management teams, the role of cascade genetic testing, pathways for arrhythmic risk stratification, and the criteria for prophylactic defibrillator implantation. Significant evidence gaps persist, particularly regarding the clinical trajectory of genetic, non-genetic and gene-elusive forms of DCM, the use of cardiovascular magnetic resonance in phenotype-negative family members with genotype-positive probands, and the development of potential aetiology-oriented therapies. Addressing these gaps could enhance clinical outcomes and inform future research directions and guideline development.
Collapse
Affiliation(s)
- Anna Sorella
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Kristian Galanti
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Lorena Iezzi
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Sabina Gallina
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, SS. Annunziata Polyclinic, Chieti 66100, Italy
- Institute for Advanced Biomedical Technologies, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Selma F Mohammed
- Department of Cardiology, Creighton University School of Medicine, Omaha, NE 68124, USA
| | - Neha Sekhri
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
- Newham University Hospital, Barts Health NHS Trust, London EC1M 6BQ, UK
- William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Mohammed Majid Akhtar
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas’ NHS Foundation Trust, London SW3 6NP, UK
| | - Sanjay K Prasad
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas’ NHS Foundation Trust, London SW3 6NP, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Choudhary Anwar Ahmed Chahal
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
- William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Center for Inherited Cardiovascular Diseases, Department of Cardiology, WellSpan Health, 30 Monument Rd, York, PA 17403, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Str, SW Rochester, MN 55905, USA
| | - Fabrizio Ricci
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, SS. Annunziata Polyclinic, Chieti 66100, Italy
- Institute for Advanced Biomedical Technologies, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Mohammed Yunus Khanji
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
- Newham University Hospital, Barts Health NHS Trust, London EC1M 6BQ, UK
- William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
6
|
Garcia-Montero M, Fanous Y, Krahn AD, Davies B, Cadrin-Tourigny J, Roberts JD. New Insights Into Genetic Right Ventricular Cardiomyopathies. Can J Cardiol 2025:S0828-282X(25)00130-8. [PMID: 39956378 DOI: 10.1016/j.cjca.2025.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/24/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025] Open
Abstract
Inherited right ventricular disease in the form of arrhythmogenic right ventricular cardiomyopathy (ARVC) was first described 40 years ago. The ARVC-causing genes have progressively been identified from the year 2000, accompanied by a robust journey of deep phenotyping. The explosion of genotype and phenotype data coupled with a collaborative spirit in the ARVC community has led to an immense advance in our understanding of the various faces of this disease, with a recent focus on gene-specific phenotypes and risk assessment and mitigation. The modern cardiogenetic team has a wealth of information that informs the biology of the disease, its phenotypic expression, and the processes of care to detect the presence and progression of disease. Gene-specific considerations will raise the bar in precision medicine applied to diagnosis, natural history, and potentially curative interventions with targeted small molecules and gene therapy. This is an exciting time for the ARVC collaborative community to usher in a new era in changing the course of ARVC for patients and their families.
Collapse
Affiliation(s)
- Marta Garcia-Montero
- Cardiovascular Genetics Centre and Electrophysiology Service, Montréal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Yehia Fanous
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Andrew D Krahn
- Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Brianna Davies
- Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julia Cadrin-Tourigny
- Cardiovascular Genetics Centre and Electrophysiology Service, Montréal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Jason D Roberts
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada; Population Health Research Institute, Hamilton, Ontario, Canada
| |
Collapse
|
7
|
Wang Z, Wu J, Lv Z, Liang P, Li Q, Li Y, Guo Y. LMNA-related cardiomyopathy: From molecular pathology to cardiac gene therapy. J Adv Res 2025:S2090-1232(25)00001-3. [PMID: 39827909 DOI: 10.1016/j.jare.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/29/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND The genetic variants of LMNA cause an array of diseases that often affect the heart. LMNA-related cardiomyopathy exhibits high-penetrance and early-onset phenotypes that lead to late-stage heart failure or lethal arrhythmia. As a subtype of dilated cardiomyopathy and arrhythmogenic cardiomyopathy, LMNA-related cardiac dysfunction is resistant to existing cardiac therapeutic strategies, leaving a major unmet clinical need in cardiomyopathy management. AIM OF REVIEW Here we comprehensively summarize current knowledge about the genetic basis, disease models and pathological mechanisms of LMNA-related cardiomyopathy. Recent translational studies were highlighted to indicate new therapeutic modalities such as gene supplementation, gene silencing and genome editing therapy, which offer potential opportunities to overcome the difficulties in the development of specific drugs for this disease. KEY SCIENTIFIC CONCEPTS OF REVIEW LMNA-related cardiomyopathy involves many diverse disease mechanisms that preclude small-molecule drugs that target only a small fraction of the mechanisms. Agreeing to this notion, the first-in-human clinical trial for this disease recently reported futility. By contrast, gene therapy offers the new hope to directly intervene LMNA variants and demonstrates a tremendous potential for breakthrough therapy for this disease. Concepts in this review are also applicable to studies of other genetic diseases that lack effective therapeutics.
Collapse
Affiliation(s)
- Ze Wang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Jiahao Wu
- Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengyuan Lv
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Ping Liang
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China.
| | - Qirui Li
- Department of Cardiology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| | - Yifei Li
- Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Yuxuan Guo
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| |
Collapse
|
8
|
Van Linthout S, Stellos K, Giacca M, Bertero E, Cannata A, Carrier L, Garcia‐Pavia P, Ghigo A, González A, Haugaa KH, Imazio M, Lopes LR, Most P, Pollesello P, Schunkert H, Streckfuss‐Bömeke K, Thum T, Tocchetti CG, Tschöpe C, van der Meer P, van Rooij E, Metra M, Rosano GM, Heymans S. State of the art and perspectives of gene therapy in heart failure. A scientific statement of the Heart Failure Association of the ESC, the ESC Council on Cardiovascular Genomics and the ESC Working Group on Myocardial & Pericardial Diseases. Eur J Heart Fail 2025; 27:5-25. [PMID: 39576264 PMCID: PMC11798634 DOI: 10.1002/ejhf.3516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/06/2024] [Accepted: 10/23/2024] [Indexed: 02/07/2025] Open
Abstract
Gene therapy has recently become a reality in the treatment of cardiovascular diseases. Strategies to modulate gene expression using antisense oligonucleotides or small interfering RNA are proving to be safe and effective in the clinic. Adeno-associated viral vector-based gene delivery and CRISPR-Cas9-based genome editing have emerged as efficient strategies for gene delivery and repair in humans. Overall, gene therapy holds the promise not only of expanding current treatment options, but also of intervening in previously untackled causal disease mechanisms with little side effects. This scientific statement provides a comprehensive overview of the various modalities of gene therapy used to treat heart failure and some of its risk factors, and their application in the clinical setting. It discusses specifically the possibilities of gene therapy for hereditary heart diseases and (non)-genetic heart failure. Furthermore, it addresses safety and clinical trial design issues and challenges for future regulatory strategies.
Collapse
Affiliation(s)
- Sophie Van Linthout
- Berlin Institute of Health (BIH) at Charité – Universitätmedizin BerlinBIH Center for Regenerative Therapies (BCRT)BerlinGermany
- German Center for Cardiovascular Research (DZHK)partner site BerlinBerlinGermany
| | - Konstantinos Stellos
- Department of Cardiovascular Research, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive CareUniversity Medical Centre Mannheim, Heidelberg UniversityMannheimGermany
- German Centre for Cardiovascular Research (DZHK)partner site Heidelberg/MannheimMannheimGermany
- Helmholtz Institute for Translational AngioCardioScience (HI‐TAC)MannheimGermany
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical SciencesNewcastle UniversityNewcastleUK
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, UK; Department of Medical SciencesUniversity of TriesteTriesteItaly
| | - Edoardo Bertero
- Cardiovascular Unit, Department of Internal MedicineUniversity of GenovaGenovaItaly
| | - Antonio Cannata
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research ExcellenceKing's College LondonLondonUK
| | - Lucie Carrier
- Department of Experimental Pharmacology and ToxicologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre for Cardiovascular Research (DZHK)partner site Hamburg/Kiel/LübeckHamburgGermany
| | - Pablo Garcia‐Pavia
- Hospital Universitario Puerta de Hierro Majadahonda, IDIPHISA, CIBERCVMadridSpain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- Universidad Francisco de Vitoria (UFV)MadridSpain
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health SciencesMolecular Biotechnology Center "Guido Tarone," University of TorinoTorinoItaly
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA and Department of Pathology, Anatomy and PhysiologyUniversidad de NavarraPamplonaSpain
- IdiSNANavarra Institute for Health ResearchPamplonaSpain
- CIBERCV (Network for Biomedical Research in Cardiovascular Disease)Instituto de Salud Carlos IIMadridSpain
| | - Kristina H. Haugaa
- ProCardio Center for Innovation, Department of CardiologyOslo University Hospital, RikshospitaletOsloNorway
- Faculty of Medicine, Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Massimo Imazio
- Department of Medicine (DMED), University of Udine, and Cardiothoracic Department ASUFCUniversity Hospital Santa Maria della MisericordiaUdineItaly
| | - Luis R. Lopes
- Institute of Cardiovascular ScienceUniversity College LondonLondonUK
- Barts Heart Centre, St Bartholomew's HospitalLondonUK
| | - Patrick Most
- Department of Cardiology, Angiology, PulmonologyUniversity Hospital HeidelbergHeidelbergGermany
| | | | - Heribert Schunkert
- Department of Cardiology, Deutsches Herzzentrum MünchenTechnische Universität MünchenMunichGermany
- German Center for Cardiovascular Research (DZHK)Partner Site Munich Heart AllianceMunichGermany
| | - Katrin Streckfuss‐Bömeke
- Clinic for Cardiology and PneumologyUniversity Medical CenterGöttingenGermany
- German Center for Cardiovascular Research (DZHK), Partner site GöttingenGöttingenGermany
- Institute of Pharmacology and ToxicologyUniversity of WürzburgWürzburgGermany
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC)University Clinic WürzburgWürzburgGermany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS)Hannover Medical SchoolHannoverGermany
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences; Center for Basic and Clinical Immunology Research (CISI); Interdepartmental Center for Clinical and Translational Research (CIRCET); Interdepartmental Hypertension Research Center (CIRIAPA)Federico II UniversityNaplesItaly
| | - Carsten Tschöpe
- Berlin Institute of Health (BIH) at Charité – Universitätmedizin BerlinBIH Center for Regenerative Therapies (BCRT)BerlinGermany
- German Center for Cardiovascular Research (DZHK)partner site BerlinBerlinGermany
- Deutsches Herzzentrum der Charité (DHZC), Department of Cardiology, Angiology and Intensive MedicineCampus Virchow KlinikumBerlinGermany
| | - Peter van der Meer
- Department of CardiologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Eva van Rooij
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center UtrechtUtrechtThe Netherlands
- Department of CardiologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Marco Metra
- Cardiology, ASST Spedali Civili di Brescia, Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | - Giuseppe M.C. Rosano
- Cardiovascular Clinical Academic Group, St. George's University Hospitals, NHS TrustUniversity of LondonLondonUK
- Cardiology, San Raffaele Cassino HospitalCassinoItaly
- Department of Human Sciences and Promotion of Quality of LifeSan Raffaele University of RomeRomeItaly
| | - Stephane Heymans
- Centre for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
- Department of CardiologyMaastricht University, CARIM School for Cardiovascular DiseasesMaastrichtThe Netherlands
- European Reference Network for Rare Low Prevalence and Complex Diseases of the Heart (ERN GUARD‐Heart)AmsterdamThe Netherlands
| |
Collapse
|
9
|
Venturiello D, Tiberi PG, Perulli F, Nardoianni G, Guida L, Barsali C, Terrone C, Cianca A, Lustri C, Sclafani M, Tini G, Barbato E, Musumeci B. Unveiling the Future of Cardiac Care: A Review of Gene Therapy in Cardiomyopathies. Int J Mol Sci 2024; 25:13147. [PMID: 39684857 DOI: 10.3390/ijms252313147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
For years, the treatment of many cardiomyopathies has been solely focused on symptom management. However, cardiomyopathies have a genetic substrate, and directing therapy towards the pathophysiology rather than the epiphenomenon of the disease may be a winning strategy. Gene therapy involves the insertion of genes or the modification of existing ones and their regulatory elements through strategies like gene replacement and gene editing. Recently, gene therapy for cardiac amyloidosis and Duchenne muscular dystrophy has received approval, and important clinical trials are currently evaluating gene therapy methods for rare heart diseases like Friedreich's Ataxia, Danon disease, Fabry disease, and Pompe Disease. Furthermore, favorable results have been noted in animal studies receiving gene therapy for hypertrophic, dilated, and arrhythmogenic cardiomyopathy. This review discusses gene therapy methods, ongoing clinical trials, and future goals in this area.
Collapse
Affiliation(s)
- Damiano Venturiello
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| | - Pier Giorgio Tiberi
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| | - Francesco Perulli
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| | - Giulia Nardoianni
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| | - Leonardo Guida
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| | - Carlo Barsali
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| | - Carlo Terrone
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| | - Alessandro Cianca
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| | - Camilla Lustri
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| | - Matteo Sclafani
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London SW3 6PY, UK
| | - Giacomo Tini
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| | - Emanuele Barbato
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| | - Beatrice Musumeci
- Cardiology, Clinical and Molecular Medicine Department, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
10
|
Loh YH, Lv J, Goh Y, Sun X, Zhu X, Muheyati M, Luan Y. Remodelling of T-Tubules and Associated Calcium Handling Dysfunction in Heart Failure: Mechanisms and Therapeutic Insights. Can J Cardiol 2024; 40:2569-2588. [PMID: 39455023 DOI: 10.1016/j.cjca.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
In cardiomyocytes, transverse tubules (T-tubules) are sarcolemmal invaginations that facilitate excitation-contraction coupling and diastolic function. The clinical significance of T-tubules has become evident in that their remodelling is recognised as a hallmark feature of heart failure (HF) and a key contributor to disrupted Ca2+ homeostasis, compromised cardiac function, and arrhythmogenesis. Further investigations have revealed that T-tubule remodelling is particularly pronounced in HF with reduced ejection fraction (HFrEF), but not in HF with preserved ejection fraction, implying that T-tubule remodelling may play a crucial pathophysiologic role in HFrEF. While research on the functional importance of T-tubules is ongoing, T-tubule remodelling has been found to be reversible. That finding has triggered a surge in studies aimed at identifying specific therapeutic approaches for HFrEF. This review discusses the functional importance of T-tubules and their microdomains, the pathophysiology of T-tubule remodelling, and the potential mechanisms of current HFrEF therapeutic approaches in reversing T-tubule alterations. We also highlight discrepancies regarding the roles of T-tubule proteins in the recovery process across studies to offer valuable insights for future research.
Collapse
Affiliation(s)
- Yi Hao Loh
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Jingyi Lv
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Yenfang Goh
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Xiangjie Sun
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Xianfeng Zhu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; Department of Critical Care Medicine, Hangzhou Ninth People's Hospital, China
| | - Muergen Muheyati
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Yi Luan
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; School of Medicine, Shaoxing University, China.
| |
Collapse
|
11
|
Li S, Withaar C, Rodrigues PG, Zijlstra SN, de Boer RA, Silljé HHW, Meems LMG. The NLRP3-inflammasome inhibitor MCC950 improves cardiac function in a HFpEF mouse model. Biomed Pharmacother 2024; 181:117711. [PMID: 39616735 DOI: 10.1016/j.biopha.2024.117711] [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: 07/10/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is posing a significant medical challenge due to its growing prevalence, high hospitalization rates and limited response to current treatment options. Accumulating evidence suggests that a comorbidity-driven systemic pro-inflammatory state, including activation of the NLRP3 inflammasome, contributes to the pathogenesis of HFpEF. This study aimed to investigate the potential cardiac protective effects of the selective NLRP3 inhibitor MCC950, in a mouse model of HFpEF. HFpEF was obtained in 18-22 months old female mice using high-fat diet (HFD) and angiotensin II (AngII) infusion. Mice developed HFpEF and comorbidities such as obesity, type 2 diabetes, and hypertension. MCC950 was added to HFD and groups were treated for four weeks until the study endpoint. MCC950 treatment resulted in lower plasma IL-18 levels (-47.3 %), illustrating target engagement. First, we observed that MCC950 treatment improved left ventricular function, demonstrated by enhanced global longitudinal strain (GLS, 3.9 %, P<0.01) and reverse peak longitudinal strain (RPLSR, +46.8 %, P<0.05). Second, MCC950 reduced cardiac hypertrophy (cardiomyocyte size -19.5 %, P<0.001) and fibrosis (-32.5 %, P<0.05), accompanied by lower expression of pro-fibrotic genes. Finally, MCC950 treatment reduced macrophage infiltration in left ventricular tissue and attenuated macrophage accumulation in visceral adipose tissue, even more as compared to caloric restriction. Overall, this suggests that NLRP3 inhibition could be a promising treatment for HFpEF patients with a pro-inflammatory profile, potentially improving heart function, systemic inflammation, and metabolic parameters.
Collapse
Affiliation(s)
- Sunhuo Li
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Coenraad Withaar
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Patricia G Rodrigues
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sietske N Zijlstra
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands; Cardiovascular Research Institute, Thorax Center, Department of Cardiology, Erasmus Medical Center, Rotterdam 3015 GD, the Netherlands
| | - Herman H W Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Laura M G Meems
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands.
| |
Collapse
|
12
|
Grisorio L, Bongianino R, Gianeselli M, Priori SG. Gene therapy for cardiac diseases: methods, challenges, and future directions. Cardiovasc Res 2024; 120:1664-1682. [PMID: 39302117 DOI: 10.1093/cvr/cvae207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 09/22/2024] Open
Abstract
Gene therapy is advancing at an unprecedented pace, and the recent success of clinical trials reinforces optimism and trust among the scientific community. Recently, the cardiac gene therapy pipeline, which had progressed more slowly than in other fields, has begun to advance, overcoming biological and technical challenges, particularly in treating genetic heart pathologies. The primary rationale behind the focus on monogenic cardiac diseases is the well-defined molecular mechanisms driving their phenotypes, directly linked to the pathogenicity of single genetic mutations. This aspect makes these conditions a remarkable example of 'genetically druggable' diseases. Unfortunately, current treatments for these life-threatening disorders are few and often poorly effective, underscoring the need to develop therapies to modulate or correct their molecular substrates. In this review we examine the latest advancements in cardiac gene therapy, discussing the pros and cons of different molecular approaches and delivery vectors, with a focus on their therapeutic application in cardiac inherited diseases. Additionally, we highlight the key factors that may enhance clinical translation, drawing insights from previous trials and the current prospects of cardiac gene therapy.
Collapse
Affiliation(s)
- Luca Grisorio
- Department of Internal Medicine, University of Pavia, Via Golgi 19, Pavia, 27100, Italy
| | - Rossana Bongianino
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Via Maugeri 10, Pavia, 27100, Italy
| | - Matteo Gianeselli
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA
| | - Silvia Giuliana Priori
- Department of Internal Medicine, University of Pavia, Via Golgi 19, Pavia, 27100, Italy
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Via Maugeri 10, Pavia, 27100, Italy
- Molecular Cardiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), C/ Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| |
Collapse
|
13
|
Yan J, Wang X, Cao P, Li Q, Wu H. Downregulation of miR-214 promotes dilated Cardiomyopathy Progression through PDE5A-Mediated cGMP regulation. Sci Rep 2024; 14:28070. [PMID: 39543318 PMCID: PMC11564883 DOI: 10.1038/s41598-024-78983-2] [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: 06/18/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is a myocardial disorder resulting in a substantial decline in cardiac function and potentially leading to heart failure. This research combines bioinformatics analysis with empirical validation to explore the roles and mechanisms of miR-214 in DCM. Using the DEseq2 R package, a total of 125 differentially expressed circulating miRNAs (DE c-miRNAs) and 784 DE genes (DEGs) were identified. Cross-analysis between target genes of DE c-miRNAs and DEGs identified 124 common genes, and protein-protein interaction analysis of common genes identified 11 hub genes. Twelve DE c-miRNAs were further verified by quantifying their levels in the serum of DCM patients and healthy individuals. miR-214 levels were significantly decreased in serum from DCM patients, positively correlated with left ventricular ejection fraction and left ventricular fractional shortening. Further analysis showed that miR-214 directly targets and negatively regulates phosphodiesterase 5 A (PDE5A). Elevated PDE5A expression reduced cGMP levels; however, using sildenafil, a PDE5A inhibitor, reversed this effect, substantiating the regulatory mechanism of miR-214 on cGMP via PDE5A. These results provide new potential targets for the diagnosis and treatment of DCM.
Collapse
Affiliation(s)
- Jingjing Yan
- Graduate School, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xinzhou Wang
- Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, 450002, China
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, 6 Dongfeng Rd, Zhengzhou, 450002, Henan, China
| | - Panxia Cao
- Graduate School, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Qiaozhi Li
- Heart Center, First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450002, China
| | - Hong Wu
- Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, 450002, China.
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, 6 Dongfeng Rd, Zhengzhou, 450002, Henan, China.
- Institute of Cardiovascular Disease, Henan University of Chinese Medicine, Zhengzhou, 450002, China.
| |
Collapse
|
14
|
Huang S, Li J, Li Q, Wang Q, Zhou X, Chen J, Chen X, Bellou A, Zhuang J, Lei L. Cardiomyopathy: pathogenesis and therapeutic interventions. MedComm (Beijing) 2024; 5:e772. [PMID: 39465141 PMCID: PMC11502724 DOI: 10.1002/mco2.772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Cardiomyopathy is a group of disease characterized by structural and functional damage to the myocardium. The etiologies of cardiomyopathies are diverse, spanning from genetic mutations impacting fundamental myocardial functions to systemic disorders that result in widespread cardiac damage. Many specific gene mutations cause primary cardiomyopathy. Environmental factors and metabolic disorders may also lead to the occurrence of cardiomyopathy. This review provides an in-depth analysis of the current understanding of the pathogenesis of various cardiomyopathies, highlighting the molecular and cellular mechanisms that contribute to their development and progression. The current therapeutic interventions for cardiomyopathies range from pharmacological interventions to mechanical support and heart transplantation. Gene therapy and cell therapy, propelled by ongoing advancements in overarching strategies and methodologies, has also emerged as a pivotal clinical intervention for a variety of diseases. The increasing number of causal gene of cardiomyopathies have been identified in recent studies. Therefore, gene therapy targeting causal genes holds promise in offering therapeutic advantages to individuals diagnosed with cardiomyopathies. Acting as a more precise approach to gene therapy, they are gradually emerging as a substitute for traditional gene therapy. This article reviews pathogenesis and therapeutic interventions for different cardiomyopathies.
Collapse
Affiliation(s)
- Shitong Huang
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Jiaxin Li
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Qiuying Li
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Qiuyu Wang
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Xianwu Zhou
- Department of Cardiovascular SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jimei Chen
- Department of Cardiovascular SurgeryGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| | - Xuanhui Chen
- Department of Medical Big Data CenterGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Abdelouahab Bellou
- Department of Emergency Medicine, Institute of Sciences in Emergency MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Emergency MedicineWayne State University School of MedicineDetroitMichiganUSA
| | - Jian Zhuang
- Department of Cardiovascular SurgeryGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| | - Liming Lei
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| |
Collapse
|
15
|
Bortolin RH, Nawar F, Park C, Trembley MA, Prondzynski M, Sweat ME, Wang P, Chen J, Lu F, Liou C, Berkson P, Keating EM, Yoshinaga D, Pavlaki N, Samenuk T, Cavazzoni CB, Sage PT, Ma Q, Whitehill RD, Abrams DJ, Carreon CK, Putra J, Alexandrescu S, Guo S, Tsai WC, Rubart M, Kubli D, Mullick AE, Bezzerides VJ, Pu WT. Antisense Oligonucleotide Therapy for Calmodulinopathy. Circulation 2024; 150:1199-1210. [PMID: 39155863 PMCID: PMC11747850 DOI: 10.1161/circulationaha.123.068111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 07/16/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Calmodulinopathies are rare inherited arrhythmia syndromes caused by dominant heterozygous variants in CALM1, CALM2, or CALM3, which each encode the identical CaM (calmodulin) protein. We hypothesized that antisense oligonucleotide (ASO)-mediated depletion of an affected calmodulin gene would ameliorate disease manifestations, whereas the other 2 calmodulin genes would preserve CaM level and function. METHODS We tested this hypothesis using human induced pluripotent stem cell-derived cardiomyocyte and mouse models of CALM1 pathogenic variants. RESULTS Human CALM1F142L/+ induced pluripotent stem cell-derived cardiomyocytes exhibited prolonged action potentials, modeling congenital long QT syndrome. CALM1 knockout or CALM1-depleting ASOs did not alter CaM protein level and normalized repolarization duration of CALM1F142L/+ induced pluripotent stem cell-derived cardiomyocytes. Similarly, an ASO targeting murine Calm1 depleted Calm1 transcript without affecting CaM protein level. This ASO alleviated drug-induced bidirectional ventricular tachycardia in Calm1N98S/+ mice without a deleterious effect on cardiac electrical or contractile function. CONCLUSIONS These results provide proof of concept that ASOs targeting individual calmodulin genes are potentially effective and safe therapies for calmodulinopathies.
Collapse
Affiliation(s)
- Raul H. Bortolin
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - Farina Nawar
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - Chaehyoung Park
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | | | | | - Mason E. Sweat
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - Peizhe Wang
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - Jiehui Chen
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - Fujian Lu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - Carter Liou
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - Paul Berkson
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - Erin M. Keating
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | | | - Nikoleta Pavlaki
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - Thomas Samenuk
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - Cecilia B. Cavazzoni
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School
| | - Qing Ma
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - Robert D. Whitehill
- Department of Pediatrics, Division of Cardiology, Emory University School of Medicine, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Dominic J. Abrams
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
- Center for Cardiovascular Genetics, Boston Children’s Hospital & Harvard Medical School, MA
| | - Chrystalle K. Carreon
- Cardiac Registry, Departments of Cardiology, Pathology, and Cardiac Surgery, Boston Children’s Hospital, Boston, MA, USA
| | - Juan Putra
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Sanda Alexandrescu
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Shuai Guo
- Wells Centre for Pediatric Research, Indiana University School of Medicine, Indianapolis, USA
| | - Wen-Chin Tsai
- Wells Centre for Pediatric Research, Indiana University School of Medicine, Indianapolis, USA
| | - Michael Rubart
- Wells Centre for Pediatric Research, Indiana University School of Medicine, Indianapolis, USA
| | - Dieter Kubli
- Ionis Pharmaceuticals, 2855 Gazelle Court Carlsbad, CA 92010 USA
| | - Adam E. Mullick
- Ionis Pharmaceuticals, 2855 Gazelle Court Carlsbad, CA 92010 USA
| | - Vassilios J. Bezzerides
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
- Center for Cardiovascular Genetics, Boston Children’s Hospital & Harvard Medical School, MA
| | - William T. Pu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
- Center for Cardiovascular Genetics, Boston Children’s Hospital & Harvard Medical School, MA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
16
|
Stege NM, de Boer RA, Makarewich CA, van der Meer P, Silljé HHW. Reassessing the Mechanisms of PLN-R14del Cardiomyopathy: From Calcium Dysregulation to S/ER Malformation. JACC Basic Transl Sci 2024; 9:1041-1052. [PMID: 39297138 PMCID: PMC11405888 DOI: 10.1016/j.jacbts.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 09/21/2024]
Abstract
The phospholamban (PLN) pathogenic gene variant, p.Arg14del (PLN-R14del), can lead to dilated and arrhythmogenic cardiomyopathy, resulting in heart failure. PLN-R14del cardiomyopathy has been conceptualized as a disease caused by sarco/endoplasmic reticulum calcium adenosine triphosphatase 2a (SERCA2a) superinhibition. However, recent studies raised controversy regarding the effect of PLN-R14del on SERCA activity and revealed a prominent role for abnormal PLN protein distribution and sarco/endoplasmic reticulum disorganization as underlying disease mechanism. Strategies targeting sarco/endoplasmic reticulum malformation may, therefore, prove more effective than SERCA activity modulation. This review reassesses the disease mechanisms of PLN-R14del cardiomyopathy and emphasizes the importance of dissecting the underlying molecular mechanisms to uncover targets for innovative treatments.
Collapse
Affiliation(s)
- Nienke M Stege
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rudolf A de Boer
- Erasmus Medical Center, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - Catherine A Makarewich
- Division of Molecular Cardiovascular Biology of the Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Herman H W Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
17
|
Arnautu DA, Cozma D, Lala IR, Arnautu SF, Tomescu MC, Andor M. Risk Assessment and Personalized Treatment Options in Inherited Dilated Cardiomyopathies: A Narrative Review. Biomedicines 2024; 12:1643. [PMID: 39200108 PMCID: PMC11351202 DOI: 10.3390/biomedicines12081643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 09/01/2024] Open
Abstract
Considering the worldwide impact of heart failure, it is crucial to develop approaches that can help us comprehend its root cause and make accurate predictions about its outcome. This is essential for lowering the suffering and death rates connected with this widespread illness. Cardiomyopathies frequently result from genetic factors, and the study of heart failure genetics is advancing quickly. Dilated cardiomyopathy (DCM) is the most prevalent kind of cardiomyopathy, encompassing both genetic and nongenetic abnormalities. It is distinguished by the enlargement of the left ventricle or both ventricles, accompanied by reduced contractility. The discovery of the molecular origins and subsequent awareness of the molecular mechanism is broadening our knowledge of DCM development. Additionally, it emphasizes the complicated nature of DCM and the necessity to formulate several different strategies to address the diverse underlying factors contributing to this disease. Genetic variants that can be transmitted from one generation to another can be a significant contributor to causing family or sporadic hereditary DCM. Genetic variants also play a significant role in determining susceptibility for acquired triggers for DCM. The genetic causes of DCM can have a large range of phenotypic expressions. It is crucial to select patients who are most probable to gain advantages from genetic testing. The purpose of this research is to emphasize the significance of identifying genetic DCM, the relationships between genotype and phenotype, risk assessment, and personalized therapy for both those affected and their relatives. This approach is expected to gain importance once treatment is guided by genotype-specific advice and disease-modifying medications.
Collapse
Affiliation(s)
- Diana-Aurora Arnautu
- Multidisciplinary Heart Research Center, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (D.-A.A.); (M.-C.T.)
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Dragos Cozma
- Department of Cardiology, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Ioan-Radu Lala
- Department of Cardiology, Western University Vasile Goldis, 310025 Arad, Romania
| | - Sergiu-Florin Arnautu
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Mirela-Cleopatra Tomescu
- Multidisciplinary Heart Research Center, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (D.-A.A.); (M.-C.T.)
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Minodora Andor
- Multidisciplinary Heart Research Center, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (D.-A.A.); (M.-C.T.)
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
18
|
Ivanova A, Kohl F, González-King Garibotti H, Chalupska R, Cvjetkovic A, Firth M, Jennbacken K, Martinsson S, Silva AM, Viken I, Wang QD, Wiseman J, Dekker N. In vivo phage display identifies novel peptides for cardiac targeting. Sci Rep 2024; 14:12177. [PMID: 38806609 PMCID: PMC11133476 DOI: 10.1038/s41598-024-62953-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
Heart failure remains a leading cause of mortality. Therapeutic intervention for heart failure would benefit from targeted delivery to the damaged heart tissue. Here, we applied in vivo peptide phage display coupled with high-throughput Next-Generation Sequencing (NGS) and identified peptides specifically targeting damaged cardiac tissue. We established a bioinformatics pipeline for the identification of cardiac targeting peptides. Hit peptides demonstrated preferential uptake by human induced pluripotent stem cell (iPSC)-derived cardiomyocytes and immortalized mouse HL1 cardiomyocytes, without substantial uptake in human liver HepG2 cells. These novel peptides hold promise for use in targeted drug delivery and regenerative strategies and open new avenues in cardiovascular research and clinical practice.
Collapse
Affiliation(s)
- Alena Ivanova
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden.
| | - Franziska Kohl
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solnavägen 1, Solna, 171 77, Stockholm, Sweden
| | - Hernán González-King Garibotti
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Renata Chalupska
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Aleksander Cvjetkovic
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Mike Firth
- Data Sciences and Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Karin Jennbacken
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Sofia Martinsson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Andreia M Silva
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Ida Viken
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - John Wiseman
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden
| | - Niek Dekker
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 431 50, Gothenburg, Sweden.
| |
Collapse
|
19
|
Chen Z, Li Z, Xu R, Xie Y, Li D, Zhao Y. Design, Synthesis, and In Vivo Evaluation of Isosteviol Derivatives as New SIRT3 Activators with Highly Potent Cardioprotective Effects. J Med Chem 2024; 67:6749-6768. [PMID: 38572607 DOI: 10.1021/acs.jmedchem.4c00345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Cardiovascular diseases (CVDs) persist as the predominant cause of mortality, urging the exploration of innovative pharmaceuticals. Mitochondrial dysfunction stands as a pivotal contributor to CVDs development. Sirtuin 3 (SIRT3), a prominent mitochondrial deacetylase known for its crucial role in protecting mitochondria against damage and dysfunction, has emerged as a promising therapeutic target for CVDs treatment. Utilizing isosteviol, a natural ent-beyerene diterpenoid, 24 derivatives were synthesized and evaluated in vivo using a zebrafish model, establishing a deduced structure-activity relationship. Among these, derivative 5v exhibited significant efficacy in doxorubicin-induced cardiomyopathy in zebrafish and murine models. Subsequent investigations revealed that 5v selectively elevated SIRT3 expression, leading to the upregulation of SOD2 and OPA1 expression, effectively preventing mitochondrial dysfunction, mitigating oxidative stress, and preserving cardiomyocyte viability. As a novel structural class of SIRT3 activators with robust therapeutic effects, 5v emerges as a promising candidate for further drug development.
Collapse
Affiliation(s)
- Zhenyu Chen
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhiyin Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Ruilong Xu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yufeng Xie
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Dehuai Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yu Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
20
|
Antonopoulos AS, Xintarakou A, Protonotarios A, Lazaros G, Miliou A, Tsioufis K, Vlachopoulos C. Imagenetics for Precision Medicine in Dilated Cardiomyopathy. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004301. [PMID: 38415367 DOI: 10.1161/circgen.123.004301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Dilated cardiomyopathy (DCM) is a common heart muscle disorder of nonischemic etiology associated with heart failure development and the risk of malignant ventricular arrhythmias and sudden cardiac death. A tailored approach to risk stratification and prevention of sudden cardiac death is required in genetic DCM given its variable presentation and phenotypic severity. Currently, advances in cardiogenetics have shed light on disease mechanisms, the complex genetic architecture of DCM, polygenic contributors to disease susceptibility and the role of environmental triggers. Parallel advances in imaging have also enhanced disease recognition and the identification of the wide spectrum of phenotypes falling under the DCM umbrella. Genotype-phenotype associations have been also established for specific subtypes of DCM, such as DSP (desmoplakin) or FLNC (filamin-C) cardiomyopathy but overall, they remain elusive and not readily identifiable. Also, despite the accumulated knowledge on disease mechanisms, certain aspects remain still unclear, such as which patients with DCM are at risk for disease progression or remission after treatment. Imagenetics, that is, the combination of imaging and genetics, is expected to further advance research in the field and contribute to precision medicine in DCM management and treatment. In the present article, we review the existing literature in the field, summarize the established knowledge and emerging data on the value of genetics and imaging in establishing genotype-phenotype associations in DCM and in clinical decision making for DCM patients.
Collapse
Affiliation(s)
- Alexios S Antonopoulos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Greece (A.S.A., A.X., G.L., A.M., K.T., C.V.)
| | - Anastasia Xintarakou
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Greece (A.S.A., A.X., G.L., A.M., K.T., C.V.)
| | - Alexandros Protonotarios
- Institute of Cardiovascular Science, University College London, United Kingdom (A.P.)
- Inherited Cardiovascular Disease Unit, St Bartholomew's Hospital, London, United Kingdom (A.P.)
| | - George Lazaros
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Greece (A.S.A., A.X., G.L., A.M., K.T., C.V.)
| | - Antigoni Miliou
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Greece (A.S.A., A.X., G.L., A.M., K.T., C.V.)
| | - Konstantinos Tsioufis
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Greece (A.S.A., A.X., G.L., A.M., K.T., C.V.)
| | - Charalambos Vlachopoulos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Greece (A.S.A., A.X., G.L., A.M., K.T., C.V.)
| |
Collapse
|
21
|
Stege NM, Eijgenraam TR, Oliveira Nunes Teixeira V, Feringa AM, Schouten EM, Kuster DW, van der Velden J, Wolters AH, Giepmans BN, Makarewich CA, Bassel-Duby R, Olson EN, de Boer RA, Silljé HH. DWORF Extends Life Span in a PLN-R14del Cardiomyopathy Mouse Model by Reducing Abnormal Sarcoplasmic Reticulum Clusters. Circ Res 2023; 133:1006-1021. [PMID: 37955153 PMCID: PMC10699510 DOI: 10.1161/circresaha.123.323304] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/18/2023] [Accepted: 11/01/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND The p.Arg14del variant of the PLN (phospholamban) gene causes cardiomyopathy, leading to severe heart failure. Calcium handling defects and perinuclear PLN aggregation have both been suggested as pathological drivers of this disease. Dwarf open reading frame (DWORF) has been shown to counteract PLN regulatory calcium handling function in the sarco/endoplasmic reticulum (S/ER). Here, we investigated the potential disease-modulating action of DWORF in this cardiomyopathy and its effects on calcium handling and PLN aggregation. METHODS We studied a PLN-R14del mouse model, which develops cardiomyopathy with similar characteristics as human patients, and explored whether cardiac DWORF overexpression could delay cardiac deterioration. To this end, R14Δ/Δ (homozygous PLN-R14del) mice carrying the DWORF transgene (R14Δ/ΔDWORFTg [R14Δ/Δ mice carrying the DWORF transgene]) were used. RESULTS DWORF expression was suppressed in hearts of R14Δ/Δ mice with severe heart failure. Restoration of DWORF expression in R14Δ/Δ mice delayed cardiac fibrosis and heart failure and increased life span >2-fold (from 8 to 18 weeks). DWORF accelerated sarcoplasmic reticulum calcium reuptake and relaxation in isolated cardiomyocytes with wild-type PLN, but in R14Δ/Δ cardiomyocytes, sarcoplasmic reticulum calcium reuptake and relaxation were already enhanced, and no differences were detected between R14Δ/Δ and R14Δ/ΔDWORFTg. Rather, DWORF overexpression delayed the appearance and formation of large pathogenic perinuclear PLN clusters. Careful examination revealed colocalization of sarcoplasmic reticulum markers with these PLN clusters in both R14Δ/Δ mice and human p.Arg14del PLN heart tissue, and hence these previously termed aggregates are comprised of abnormal organized S/ER. This abnormal S/ER organization in PLN-R14del cardiomyopathy contributes to cardiomyocyte cell loss and replacement fibrosis, consequently resulting in cardiac dysfunction. CONCLUSIONS Disorganized S/ER is a major characteristic of PLN-R14del cardiomyopathy in humans and mice and results in cardiomyocyte death. DWORF overexpression delayed PLN-R14del cardiomyopathy progression and extended life span in R14Δ/Δ mice, by reducing abnormal S/ER clusters.
Collapse
Affiliation(s)
- Nienke M. Stege
- Department of Cardiology, University Medical Center Groningen, University of Groningen, the Netherlands (N.M.S., T.R.E., V.O.N.T., A.M.F., E.M.S., R.A.d.B., H.H.W.S.)
| | - Tim R. Eijgenraam
- Department of Cardiology, University Medical Center Groningen, University of Groningen, the Netherlands (N.M.S., T.R.E., V.O.N.T., A.M.F., E.M.S., R.A.d.B., H.H.W.S.)
| | - Vivian Oliveira Nunes Teixeira
- Department of Cardiology, University Medical Center Groningen, University of Groningen, the Netherlands (N.M.S., T.R.E., V.O.N.T., A.M.F., E.M.S., R.A.d.B., H.H.W.S.)
| | - Anna M. Feringa
- Department of Cardiology, University Medical Center Groningen, University of Groningen, the Netherlands (N.M.S., T.R.E., V.O.N.T., A.M.F., E.M.S., R.A.d.B., H.H.W.S.)
| | - Elisabeth M. Schouten
- Department of Cardiology, University Medical Center Groningen, University of Groningen, the Netherlands (N.M.S., T.R.E., V.O.N.T., A.M.F., E.M.S., R.A.d.B., H.H.W.S.)
| | - Diederik W.D. Kuster
- Department of Physiology (D.W.D.K., J.v.d.V.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias (D.W.D.K., J.v.d.V.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - Jolanda van der Velden
- Department of Physiology (D.W.D.K., J.v.d.V.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias (D.W.D.K., J.v.d.V.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - Anouk H.G. Wolters
- Biomedical Sciences of Cells and Systems, UMC Groningen, University of Groningen, the Netherlands (A.H.G.W., B.N.G.G.)
| | - Ben N.G. Giepmans
- Biomedical Sciences of Cells and Systems, UMC Groningen, University of Groningen, the Netherlands (A.H.G.W., B.N.G.G.)
| | - Catherine A. Makarewich
- Division of Molecular Cardiovascular Biology of the Heart Institute, Cincinnati Children’s Hospital Medical Center, OH (C.A.M.)
- Department of Pediatrics, University of Cincinnati College of Medicine, OH (C.A.M.)
| | - Rhonda Bassel-Duby
- Department of Cardiology, University Medical Center Groningen, University of Groningen, the Netherlands (N.M.S., T.R.E., V.O.N.T., A.M.F., E.M.S., R.A.d.B., H.H.W.S.)
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas (R.B.-D., E.N.O.)
| | - Eric N. Olson
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas (R.B.-D., E.N.O.)
| | - Rudolf A. de Boer
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands (R.A.d.B.)
| | - Herman H.W. Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, the Netherlands (N.M.S., T.R.E., V.O.N.T., A.M.F., E.M.S., R.A.d.B., H.H.W.S.)
| |
Collapse
|
22
|
Wang S, Zhang Z, He J, Liu J, Guo X, Chu H, Xu H, Wang Y. Comprehensive review on gene mutations contributing to dilated cardiomyopathy. Front Cardiovasc Med 2023; 10:1296389. [PMID: 38107262 PMCID: PMC10722203 DOI: 10.3389/fcvm.2023.1296389] [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: 09/18/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the most common primary myocardial diseases. However, to this day, it remains an enigmatic cardiovascular disease (CVD) characterized by ventricular dilatation, which leads to myocardial contractile dysfunction. It is the most common cause of chronic congestive heart failure and the most frequent indication for heart transplantation in young individuals. Genetics and various other factors play significant roles in the progression of dilated cardiomyopathy, and variants in more than 50 genes have been associated with the disease. However, the etiology of a large number of cases remains elusive. Numerous studies have been conducted on the genetic causes of dilated cardiomyopathy. These genetic studies suggest that mutations in genes for fibronectin, cytoskeletal proteins, and myosin in cardiomyocytes play a key role in the development of DCM. In this review, we provide a comprehensive description of the genetic basis, mechanisms, and research advances in genes that have been strongly associated with DCM based on evidence-based medicine. We also emphasize the important role of gene sequencing in therapy for potential early diagnosis and improved clinical management of DCM.
Collapse
Affiliation(s)
- Shipeng Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zhiyu Zhang
- Department of Cardiovascular Medicine, The Second People's Hospital of Yibin, Yibin, China
| | - Jiahuan He
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Junqian Liu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xia Guo
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Haoxuan Chu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Hanchi Xu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yushi Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Javed S, Halliday BP. Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift? CAMBRIDGE PRISMS. PRECISION MEDICINE 2023; 1:e34. [PMID: 38550947 PMCID: PMC10953759 DOI: 10.1017/pcm.2023.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 02/06/2025]
Abstract
Precision medicine for cardiomyopathies holds great promise to improve patient outcomes costs by shifting the focus to patient-specific treatment decisions, maximising the use of therapies most likely to lead to benefit and minimising unnecessary intervention. Dilated cardiomyopathy (DCM), characterised by left ventricular dilatation and impairment, is a major cause of heart failure globally. Advances in genomic medicine have increased our understanding of the genetic architecture of DCM. Understanding the functional implications of genetic variation to reveal genotype-specific disease mechanisms is the subject of intense investigation, with advanced cardiac imaging and mutliomics approaches playing important roles. This may lead to increasing use of novel, targeted therapy. Individualised treatment and risk stratification is however made more complex by the modifying effects of common genetic variation and acquired environmental factors that help explain the variable expressivity of rare genetic variants and gene elusive disease. The next frontier must be expanding work into early disease to understand the mechanisms that drive disease expression, so that the focus can be placed on disease prevention rather than management of later symptomatic disease. Overcoming these challenges holds the key to enabling a paradigm shift in care from the management of symptomatic heart failure to prevention of disease.
Collapse
Affiliation(s)
- Saad Javed
- National Heart and Lung Institute, Imperial College London, UK
- Cardiovascular Research Centre, Cardiovascular Magnetic Resonance Unit & Inherited Cardiac Conditions Care Group, Royal Brompton and Harefield Hospitals, Part of Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Brian P. Halliday
- National Heart and Lung Institute, Imperial College London, UK
- Cardiovascular Research Centre, Cardiovascular Magnetic Resonance Unit & Inherited Cardiac Conditions Care Group, Royal Brompton and Harefield Hospitals, Part of Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| |
Collapse
|
24
|
Palmieri G, D’Ambrosio MF, Correale M, Brunetti ND, Santacroce R, Iacoviello M, Margaglione M. The Role of Genetics in the Management of Heart Failure Patients. Int J Mol Sci 2023; 24:15221. [PMID: 37894902 PMCID: PMC10607512 DOI: 10.3390/ijms242015221] [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: 09/15/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Over the last decades, the relevance of genetics in cardiovascular diseases has expanded, especially in the context of cardiomyopathies. Its relevance extends to the management of patients diagnosed with heart failure (HF), given its capacity to provide invaluable insights into the etiology of cardiomyopathies and identify individuals at a heightened risk of poor outcomes. Notably, the identification of an etiological genetic variant necessitates a comprehensive evaluation of the family lineage of the affected patients. In the future, these genetic variants hold potential as therapeutic targets with the capability to modify gene expression. In this complex setting, collaboration among cardiologists, specifically those specializing in cardiomyopathies and HF, and geneticists becomes paramount to improving individual and family health outcomes, as well as therapeutic clinical results. This review is intended to offer geneticists and cardiologists an updated perspective on the value of genetic research in HF and its implications in clinical practice.
Collapse
Affiliation(s)
- Gianpaolo Palmieri
- School of Cardiology, Department of Medical and Surgical Sciences, University of Foggia, 70122 Foggia, Italy; (G.P.); (M.C.); (N.D.B.)
| | - Maria Francesca D’Ambrosio
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.F.D.); (R.S.); (M.M.)
| | - Michele Correale
- School of Cardiology, Department of Medical and Surgical Sciences, University of Foggia, 70122 Foggia, Italy; (G.P.); (M.C.); (N.D.B.)
| | - Natale Daniele Brunetti
- School of Cardiology, Department of Medical and Surgical Sciences, University of Foggia, 70122 Foggia, Italy; (G.P.); (M.C.); (N.D.B.)
| | - Rosa Santacroce
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.F.D.); (R.S.); (M.M.)
| | - Massimo Iacoviello
- University Cardiology Unit, Polyclinic Hospital of Bari, 70124 Bari, Italy
| | - Maurizio Margaglione
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.F.D.); (R.S.); (M.M.)
| |
Collapse
|
25
|
Heymans S, Lakdawala NK, Tschöpe C, Klingel K. Dilated cardiomyopathy: causes, mechanisms, and current and future treatment approaches. Lancet 2023; 402:998-1011. [PMID: 37716772 DOI: 10.1016/s0140-6736(23)01241-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/20/2023] [Accepted: 06/13/2023] [Indexed: 09/18/2023]
Abstract
Dilated cardiomyopathy is conventionally defined as the presence of left ventricular or biventricular dilatation or systolic dysfunction in the absence of abnormal loading conditions (eg, primary valve disease) or significant coronary artery disease sufficient to cause ventricular remodelling. This definition has been recognised as overly restrictive, as left ventricular hypokinesis without dilation could be the initial presentation of dilated cardiomyopathy. The causes of dilated cardiomyopathy comprise genetic (primary dilated cardiomyopathy) or acquired factors (secondary dilated cardiomyopathy). Acquired factors include infections, toxins, cancer treatment, endocrinopathies, pregnancy, tachyarrhythmias, and immune-mediated diseases. 5-15% of patients with acquired dilated cardiomyopathy harbour a likely pathogenic or pathogenic gene variant (ie, gene mutation). Therefore, the diagnostic tests and therapeutic approach should always consider both genetic and acquired factors. This Seminar will focus on the current multidimensional diagnostic and therapeutic approach and discuss the underlying pathophysiology that could drive future treatments aiming to repair or replace the existing gene mutation, or target the specific inflammatory, metabolic, or pro-fibrotic drivers of genetic or acquired dilated cardiomyopathy.
Collapse
Affiliation(s)
- Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht, University of Maastricht & Maastricht University Medical Centre, Maastricht, Netherlands; Department of Cardiovascular Sciences, Centre for Vascular and Molecular Biology, KU Leuven, Leuven, Belgium
| | - Neal K Lakdawala
- Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carsten Tschöpe
- Department of Cardiology, Angiology, and Intensive Medicine (CVK), German Heart Center of the Charité (DHZC), Charité Universitätsmedizin, Berlin, Germany; Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
26
|
Ryan T, Roberts JD. Emerging Targeted Therapies for Inherited Cardiomyopathies and Arrhythmias. Card Electrophysiol Clin 2023; 15:261-271. [PMID: 37558297 DOI: 10.1016/j.ccep.2023.04.006] [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: 08/11/2023]
Abstract
Inherited cardiomyopathy and arrhythmia syndromes are associated with significant morbidity and mortality, particularly in young people. Medical management of these conditions has primarily been limited to agents previously developed for more common forms of heart disease and not tailored to their distinct pathophysiology. As our understanding of their underlying genetics and disease mechanisms has improved, an era of targeted therapies for these rare conditions has begun to emerge. In recent years, several novel agents have been developed and tested in preclinical models and, in some cases, have advanced to both the clinical trial and clinical approval stages with exciting results. These new treatments are derived from multiple classes of therapeutics, including small molecules, antisense oligonucleotides, small interfering RNAs, adeno-associated virus-mediated gene therapies, and in vivo gene editing. Collectively, they carry the promise of revolutionizing management of affected patients and their families.
Collapse
Affiliation(s)
- Tammy Ryan
- McMaster University, Hamilton, Ontario, Canada; Department of Medicine, Division of Cardiology, DBCVSRI, Hamilton General Hospital, Room C3-121, 237 Barton Street East, Hamilton, Ontario L8L2X2, Canada
| | - Jason D Roberts
- McMaster University, Hamilton, Ontario, Canada; DBCVSRI, Room C3-111, 237 Barton Street East, Hamilton, Ontario L8L2X2, Canada; Population Health Research Institute and Hamilton Health Sciences, Hamilton, Ontario, Canada.
| |
Collapse
|
27
|
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
|
28
|
Giacca M. Fulfilling the Promise of RNA Therapies for Cardiac Repair and Regeneration. Stem Cells Transl Med 2023; 12:527-535. [PMID: 37440203 PMCID: PMC10427962 DOI: 10.1093/stcltm/szad038] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/07/2023] [Indexed: 07/14/2023] Open
Abstract
The progressive appreciation that multiple types of RNAs regulate virtually all aspects of tissue function and the availability of effective tools to deliver RNAs in vivo now offers unprecedented possibilities for obtaining RNA-based therapeutics. For the heart, RNA therapies can be developed that stimulate endogenous repair after cardiac damage. Applications in this area include acute cardioprotection after ischemia or cancer chemotherapy, therapeutic angiogenesis to promote new blood vessel formation, regeneration to form new cardiac mass, and editing of mutations to cure inherited cardiac disease. While the potential of RNA therapeutics for all these conditions is exciting, the field is still in its infancy. A number of roadblocks need to be overcome for RNA therapies to become effective, in particular, related to the problem of delivering RNA medicines into the cells and targeting them specifically to the heart.
Collapse
Affiliation(s)
- Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King’s College London, London, UK
- Department of Medical Sciences, University of Trieste, Italy
| |
Collapse
|
29
|
de Boer RA. Should we offer preventive treatment to all carriers of PLN p.(Arg14del) variant? : Pro: Provide pre-emptive treatment to asymptomatic carriers. Neth Heart J 2023:10.1007/s12471-023-01793-0. [PMID: 37493902 PMCID: PMC10400733 DOI: 10.1007/s12471-023-01793-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2023] [Indexed: 07/27/2023] Open
Affiliation(s)
- Rudolf A de Boer
- Department of Cardiology, Erasmus University Medical Centre, Rotterdam, The Netherlands.
| |
Collapse
|
30
|
Doevendans PA. Should we offer preventive treatment to all carriers of PLN p.(Arg14del) variant? : Con: Do no harm to asymptomatic carriers. Neth Heart J 2023:10.1007/s12471-023-01794-z. [PMID: 37491506 PMCID: PMC10400497 DOI: 10.1007/s12471-023-01794-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2023] [Indexed: 07/27/2023] Open
Affiliation(s)
- Pieter A Doevendans
- University Medical Centre Utrecht, Central Military Hospital, Utrecht, The Netherlands.
- Netherlands Heart Institute, Utrecht, The Netherlands.
| |
Collapse
|
31
|
Lira JR, Guymon AL, Yang L, Sternburg JO, Giri S, Wang X. The double-hit protocol induces HFpEF and impairs myocardial ubiquitin-proteasome system performance in FVB/N mice. Front Physiol 2023; 14:1208153. [PMID: 37362441 PMCID: PMC10285383 DOI: 10.3389/fphys.2023.1208153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a leading cause of death and disability, with its prevalence surpassing that of heart failure with reduced ejection fraction. Obesity and hypertension are often associated with HFpEF. HFpEF can be modeled through simultaneous metabolic and hypertensive stresses in male C57BL/6N mice provoked by a combination treatment of a high-fat diet (HFD) and constitutive nitric oxide synthase inhibition by Nω-nitro-L-arginine methyl-ester (L-NAME). Ubiquitin-proteasome system (UPS) dysfunction was detected in many forms of cardiomyopathy, but whether it occurs in HFpEF remains unknown. We report successful modeling of HFpEF in male FVB/N mice and, by taking advantage of a transgenic UPS reporter mouse, we have detected myocardial UPS functioning impairment during HFpEF, suggesting a pathogenic role for impaired protein degradation in the development and progression of HFpEF.
Collapse
|
32
|
Skogestad J, Albert I, Hougen K, Lothe GB, Lunde M, Eken OS, Veras I, Huynh NTT, Børstad M, Marshall S, Shen X, Louch WE, Robinson EL, Cleveland JC, Ambardekar AV, Schwisow JA, Jonas E, Calejo AI, Morth JP, Taskén K, Melleby AO, Lunde PK, Sjaastad I, Carlson CR, Aronsen JM. Disruption of Phosphodiesterase 3A Binding to SERCA2 Increases SERCA2 Activity and Reduces Mortality in Mice With Chronic Heart Failure. Circulation 2023; 147:1221-1236. [PMID: 36876489 DOI: 10.1161/circulationaha.121.054168] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 02/08/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Increasing SERCA2 (sarco[endo]-plasmic reticulum Ca2+ ATPase 2) activity is suggested to be beneficial in chronic heart failure, but no selective SERCA2-activating drugs are available. PDE3A (phosphodiesterase 3A) is proposed to be present in the SERCA2 interactome and limit SERCA2 activity. Disruption of PDE3A from SERCA2 might thus be a strategy to develop SERCA2 activators. METHODS Confocal microscopy, 2-color direct stochastic optical reconstruction microscopy, proximity ligation assays, immunoprecipitations, peptide arrays, and surface plasmon resonance were used to investigate colocalization between SERCA2 and PDE3A in cardiomyocytes, map the SERCA2/PDE3A interaction sites, and optimize disruptor peptides that release PDE3A from SERCA2. Functional experiments assessing the effect of PDE3A-binding to SERCA2 were performed in cardiomyocytes and HEK293 vesicles. The effect of SERCA2/PDE3A disruption by the disruptor peptide OptF (optimized peptide F) on cardiac mortality and function was evaluated during 20 weeks in 2 consecutive randomized, blinded, and controlled preclinical trials in a total of 148 mice injected with recombinant adeno-associated virus 9 (rAAV9)-OptF, rAAV9-control (Ctrl), or PBS, before undergoing aortic banding (AB) or sham surgery and subsequent phenotyping with serial echocardiography, cardiac magnetic resonance imaging, histology, and functional and molecular assays. RESULTS PDE3A colocalized with SERCA2 in human nonfailing, human failing, and rodent myocardium. Amino acids 277-402 of PDE3A bound directly to amino acids 169-216 within the actuator domain of SERCA2. Disruption of PDE3A from SERCA2 increased SERCA2 activity in normal and failing cardiomyocytes. SERCA2/PDE3A disruptor peptides increased SERCA2 activity also in the presence of protein kinase A inhibitors and in phospholamban-deficient mice, and had no effect in mice with cardiomyocyte-specific inactivation of SERCA2. Cotransfection of PDE3A reduced SERCA2 activity in HEK293 vesicles. Treatment with rAAV9-OptF reduced cardiac mortality compared with rAAV9-Ctrl (hazard ratio, 0.26 [95% CI, 0.11 to 0.63]) and PBS (hazard ratio, 0.28 [95% CI, 0.09 to 0.90]) 20 weeks after AB. Mice injected with rAAV9-OptF had improved contractility and no difference in cardiac remodeling compared with rAAV9-Ctrl after aortic banding. CONCLUSIONS Our results suggest that PDE3A regulates SERCA2 activity through direct binding, independently of the catalytic activity of PDE3A. Targeting the SERCA2/PDE3A interaction prevented cardiac mortality after AB, most likely by improving cardiac contractility.
Collapse
Affiliation(s)
- Jonas Skogestad
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
| | - Ingrid Albert
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
| | - Karina Hougen
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
| | - Gustav B Lothe
- Department of Pharmacology, Oslo University Hospital, Norway (G.B.L.)
- Bjørknes College, Oslo, Norway (G.B.L., J.M.A.)
| | - Marianne Lunde
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
| | - Olav Søvik Eken
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
- Department of Molecular Medicine, University of Oslo, Norway (O.S.E., I.V., N.T.T.-H., A.O.M., J.M.A.)
| | - Ioanni Veras
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
- Department of Molecular Medicine, University of Oslo, Norway (O.S.E., I.V., N.T.T.-H., A.O.M., J.M.A.)
| | - Ngoc Trang Thi Huynh
- Department of Molecular Medicine, University of Oslo, Norway (O.S.E., I.V., N.T.T.-H., A.O.M., J.M.A.)
| | - Mira Børstad
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
| | - Serena Marshall
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
| | - Xin Shen
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
| | - Emma Louise Robinson
- Division of Cardiology, Department of Medicine (E.L.R., A.V.A., J.A.S., E.J.), University of Colorado Anschutz Medical Campus, Aurora
| | - Joseph C Cleveland
- Department of Surgery (J.C.C.), University of Colorado Anschutz Medical Campus, Aurora
| | - Amrut V Ambardekar
- Division of Cardiology, Department of Medicine (E.L.R., A.V.A., J.A.S., E.J.), University of Colorado Anschutz Medical Campus, Aurora
| | - Jessica A Schwisow
- Division of Cardiology, Department of Medicine (E.L.R., A.V.A., J.A.S., E.J.), University of Colorado Anschutz Medical Campus, Aurora
| | - Eric Jonas
- Division of Cardiology, Department of Medicine (E.L.R., A.V.A., J.A.S., E.J.), University of Colorado Anschutz Medical Campus, Aurora
| | - Ana I Calejo
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership (A.I.C.C., J.P.M., K.T.), Oslo University Hospital and University of Oslo, Norway
| | - Jens Preben Morth
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership (A.I.C.C., J.P.M., K.T.), Oslo University Hospital and University of Oslo, Norway
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby (J.P.M.)
| | - Kjetil Taskén
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership (A.I.C.C., J.P.M., K.T.), Oslo University Hospital and University of Oslo, Norway
- Institute for Cancer Research, Oslo University Hospital and Institute for Clinical Medicine, University of Oslo, Norway (K.T.)
| | - Arne Olav Melleby
- Department of Molecular Medicine, University of Oslo, Norway (O.S.E., I.V., N.T.T.-H., A.O.M., J.M.A.)
| | - Per Kristian Lunde
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
| | - Cathrine Rein Carlson
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research (J.S., I.A., K.H., M.L., O.S.E., I.V., M.B., S.M., X.S., W.E.L., P.K.L., I.S., C.R.C., J.M.A.), Oslo University Hospital and University of Oslo, Norway
- Bjørknes College, Oslo, Norway (G.B.L., J.M.A.)
- Department of Molecular Medicine, University of Oslo, Norway (O.S.E., I.V., N.T.T.-H., A.O.M., J.M.A.)
| |
Collapse
|
33
|
Vafiadaki E, Glijnis PC, Doevendans PA, Kranias EG, Sanoudou D. Phospholamban R14del disease: The past, the present and the future. Front Cardiovasc Med 2023; 10:1162205. [PMID: 37144056 PMCID: PMC10151546 DOI: 10.3389/fcvm.2023.1162205] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/03/2023] [Indexed: 05/06/2023] Open
Abstract
Arrhythmogenic cardiomyopathy affects significant number of patients worldwide and is characterized by life-threatening ventricular arrhythmias and sudden cardiac death. Mutations in multiple genes with diverse functions have been reported to date including phospholamban (PLN), a key regulator of sarcoplasmic reticulum (SR) Ca2+ homeostasis and cardiac contractility. The PLN-R14del variant in specific is recognized as the cause in an increasing number of patients worldwide, and extensive investigations have enabled rapid advances towards the delineation of PLN-R14del disease pathogenesis and discovery of an effective treatment. We provide a critical overview of current knowledge on PLN-R14del disease pathophysiology, including clinical, animal model, cellular and biochemical studies, as well as diverse therapeutic approaches that are being pursued. The milestones achieved in <20 years, since the discovery of the PLN R14del mutation (2006), serve as a paradigm of international scientific collaboration and patient involvement towards finding a cure.
Collapse
Affiliation(s)
- Elizabeth Vafiadaki
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Correspondence: Elizabeth Vafiadaki Despina Sanoudou
| | - Pieter C. Glijnis
- Stichting Genetische Hartspierziekte PLN, Phospholamban Foundation, Wieringerwerf, Netherlands
| | - Pieter A. Doevendans
- Netherlands Heart Institute, Utrecht, Netherlands
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Evangelia G. Kranias
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Despina Sanoudou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Correspondence: Elizabeth Vafiadaki Despina Sanoudou
| |
Collapse
|
34
|
Giacca M. Gene editing for cardiomyopathy takes a step forward. Cardiovasc Res 2022; 118:3011-3012. [PMID: 36223597 PMCID: PMC9732510 DOI: 10.1093/cvr/cvac164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 10/06/2022] [Indexed: 12/15/2022] Open
Affiliation(s)
- Mauro Giacca
- King’s College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK
| |
Collapse
|
35
|
Dong M, Liu J, Liu C, Wang H, Sun W, Liu B. CRISPR/CAS9: A promising approach for the research and treatment of cardiovascular diseases. Pharmacol Res 2022; 185:106480. [PMID: 36191879 DOI: 10.1016/j.phrs.2022.106480] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 10/31/2022]
Abstract
The development of gene-editing technology has been one of the biggest advances in biomedicine over the past two decades. Not only can it be used as a research tool to build a variety of disease models for the exploration of disease pathogenesis at the genetic level, it can also be used for prevention and treatment. This is done by intervening with the expression of target genes and carrying out precise molecular targeted therapy for diseases. The simple and flexible clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene-editing technology overcomes the limitations of zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs). For this reason, it has rapidly become a preferred method for gene editing. As a new gene intervention method, CRISPR/Cas9 has been widely used in the clinical treatment of tumours and rare diseases; however, its application in the field of cardiovascular diseases is currently limited. This article reviews the application of the CRISPR/Cas9 editing technology in cardiovascular disease research and treatment, and discusses the limitations and prospects of this technology.
Collapse
Affiliation(s)
- Mengying Dong
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Jiangen Liu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Caixia Liu
- Department of Neurology, The Liaoning Province People's Hospital, 33 Wenyi Road, ShenYang, China, 110016
| | - He Wang
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Wei Sun
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041.
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041.
| |
Collapse
|
36
|
Deiman FE, Bomer N, van der Meer P, Grote Beverborg N. Review: Precision Medicine Approaches for Genetic Cardiomyopathy: Targeting Phospholamban R14del. Curr Heart Fail Rep 2022; 19:170-179. [PMID: 35699837 PMCID: PMC9329159 DOI: 10.1007/s11897-022-00558-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE OF REVIEW Heart failure is a syndrome with poor prognosis and no curative options for the majority of patients. The standard one-size-fits-all-treatment approach, targeting neurohormonal dysregulations, helps to modulate symptoms of heart failure, but fails to address the cause of the problem. Precision medicine aims to go beyond symptom modulation and targets pathophysiological mechanisms that underlie disease. In this review, an overview of how precision medicine can be approached as a treatment strategy for genetic heart disease will be discussed. PLN R14del, a genetic mutation known to cause cardiomyopathy, will be used as an example to describe the potential and pitfalls of precision medicine. RECENT FINDINGS PLN R14del is characterized by several disease hallmarks including calcium dysregulation, metabolic dysfunction, and protein aggregation. The identification of disease-related biological pathways and the effective targeting using several modalities, including gene silencing and signal transduction modulation, may eventually provide novel treatments for genetic heart disease. We propose a workflow on how to approach precision medicine in heart disease. This workflow focuses on deep phenotyping of patient derived material, including in vitro disease modeling. This will allow identification of therapeutic targets and disease modifiers, to be used for the identification of novel biomarkers and the development of precision medicine approaches for genetic cardiomyopathies.
Collapse
Affiliation(s)
- Frederik E Deiman
- Department of Cardiology, University Medical Center Groningen, University of Groningen, UMCG Post-zone AB43, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, UMCG Post-zone AB43, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, UMCG Post-zone AB43, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Niels Grote Beverborg
- Department of Cardiology, University Medical Center Groningen, University of Groningen, UMCG Post-zone AB43, PO Box 30.001, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
37
|
Tabata T, Kuramoto Y, Ohtani T, Miyawaki H, Miyashita Y, Sera F, Kioka H, Higo S, Asano Y, Hikoso S, Sakata Y. Phospholamban p.Arg14del Cardiomyopathy: A Japanese Case Series. Intern Med 2022; 61:1987-1993. [PMID: 34924461 PMCID: PMC9334245 DOI: 10.2169/internalmedicine.8594-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Phospholamban p.Arg14del is reported to cause hereditary cardiomyopathy with malignant ventricular tachycardia (VT) and advanced heart failure. However, the clinical courses of Japanese cardiomyopathy patients with phospholamban p.Arg14del remain uncharacterized. We identified five patients with this variant. All patients were diagnosed with dilated cardiomyopathy (DCM), developed end-stage heart failure and experienced VT requiring implantable cardioverter defibrillator discharge. Four patients survived after implantation of a left ventricular assist device (LVAD), while one patient who refused LVAD implantation died of heart failure. Based on the severe course of the disease, we propose genetic screening for phospholamban p.Arg14del in DCM patients.
Collapse
Affiliation(s)
- Tomoka Tabata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Japan
| | - Yuki Kuramoto
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Japan
| | - Tomohito Ohtani
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Japan
| | - Hiroshi Miyawaki
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Japan
| | - Yohei Miyashita
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Japan
| | - Fusako Sera
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Japan
| | - Hidetaka Kioka
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Japan
| | - Shuichiro Higo
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Japan
| | - Shungo Hikoso
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Japan
| |
Collapse
|
38
|
Blocking phospholamban with VHH intrabodies enhances contractility and relaxation in heart failure. Nat Commun 2022; 13:3018. [PMID: 35641497 PMCID: PMC9156741 DOI: 10.1038/s41467-022-29703-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 03/28/2022] [Indexed: 12/19/2022] Open
Abstract
The dysregulated physical interaction between two intracellular membrane proteins, the sarco/endoplasmic reticulum Ca2+ ATPase and its reversible inhibitor phospholamban, induces heart failure by inhibiting calcium cycling. While phospholamban is a bona-fide therapeutic target, approaches to selectively inhibit this protein remain elusive. Here, we report the in vivo application of intracellular acting antibodies (intrabodies), derived from the variable domain of camelid heavy-chain antibodies, to modulate the function of phospholamban. Using a synthetic VHH phage-display library, we identify intrabodies with high affinity and specificity for different conformational states of phospholamban. Rapid phenotypic screening, via modified mRNA transfection of primary cells and tissue, efficiently identifies the intrabody with most desirable features. Adeno-associated virus mediated delivery of this intrabody results in improvement of cardiac performance in a murine heart failure model. Our strategy for generating intrabodies to investigate cardiac disease combined with modified mRNA and adeno-associated virus screening could reveal unique future therapeutic opportunities. Here the authors use modified RNA and VHH libraries to generate intrabodies that target dysregulated interactions between two calcium handling proteins in failing cardiomyocytes. Heart specific expression of the intrabodies in a murine heart failure model results in improved cardiac function.
Collapse
|
39
|
Signaling cascades in the failing heart and emerging therapeutic strategies. Signal Transduct Target Ther 2022; 7:134. [PMID: 35461308 PMCID: PMC9035186 DOI: 10.1038/s41392-022-00972-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic heart failure is the end stage of cardiac diseases. With a high prevalence and a high mortality rate worldwide, chronic heart failure is one of the heaviest health-related burdens. In addition to the standard neurohormonal blockade therapy, several medications have been developed for chronic heart failure treatment, but the population-wide improvement in chronic heart failure prognosis over time has been modest, and novel therapies are still needed. Mechanistic discovery and technical innovation are powerful driving forces for therapeutic development. On the one hand, the past decades have witnessed great progress in understanding the mechanism of chronic heart failure. It is now known that chronic heart failure is not only a matter involving cardiomyocytes. Instead, chronic heart failure involves numerous signaling pathways in noncardiomyocytes, including fibroblasts, immune cells, vascular cells, and lymphatic endothelial cells, and crosstalk among these cells. The complex regulatory network includes protein-protein, protein-RNA, and RNA-RNA interactions. These achievements in mechanistic studies provide novel insights for future therapeutic targets. On the other hand, with the development of modern biological techniques, targeting a protein pharmacologically is no longer the sole option for treating chronic heart failure. Gene therapy can directly manipulate the expression level of genes; gene editing techniques provide hope for curing hereditary cardiomyopathy; cell therapy aims to replace dysfunctional cardiomyocytes; and xenotransplantation may solve the problem of donor heart shortages. In this paper, we reviewed these two aspects in the field of failing heart signaling cascades and emerging therapeutic strategies based on modern biological techniques.
Collapse
|
40
|
Hulsurkar MM, Lahiri SK, Karch J, Wang MC, Wehrens XHT. Targeting calcium-mediated inter-organellar crosstalk in cardiac diseases. Expert Opin Ther Targets 2022; 26:303-317. [PMID: 35426759 PMCID: PMC9081256 DOI: 10.1080/14728222.2022.2067479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/14/2022] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Abnormal calcium signaling between organelles such as the sarcoplasmic reticulum (SR), mitochondria and lysosomes is a key feature of heart diseases. Calcium serves as a secondary messenger mediating inter-organellar crosstalk, essential for maintaining the cardiomyocyte function. AREAS COVERED This article examines the available literature related to calcium channels and transporters involved in inter-organellar calcium signaling. The SR calcium-release channels ryanodine receptor type-2 (RyR2) and inositol 1,4,5-trisphosphate receptor (IP3R), and calcium-transporter SR/ER-ATPase 2a (SERCA2a) are illuminated. The roles of mitochondrial voltage-dependent anion channels (VDAC), the mitochondria Ca2+ uniporter complex (MCUC), and the lysosomal H+/Ca2+ exchanger, two pore channels (TPC), and transient receptor potential mucolipin (TRPML) are discussed. Furthermore, recent studies showing calcium-mediated crosstalk between the SR, mitochondria, and lysosomes as well as how this crosstalk is dysregulated in cardiac diseases are placed under the spotlight. EXPERT OPINION Enhanced SR calcium release via RyR2 and reduced SR reuptake via SERCA2a, increased VDAC and MCUC-mediated calcium uptake into mitochondria, and enhanced lysosomal calcium-release via lysosomal TPC and TRPML may all contribute to aberrant calcium homeostasis causing heart disease. While mechanisms of this crosstalk need to be studied further, interventions targeting these calcium channels or combinations thereof might represent a promising therapeutic strategy.
Collapse
Affiliation(s)
- Mohit M Hulsurkar
- Baylor College of Medicine, Houston TX USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Satadru K Lahiri
- Baylor College of Medicine, Houston TX USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Jason Karch
- Baylor College of Medicine, Houston TX USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Meng C Wang
- Baylor College of Medicine, Houston TX USA
- Huffington Center on Aging, Baylor College of Medicine, Houston TX USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Xander H T Wehrens
- Baylor College of Medicine, Houston TX USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine (Cardiology), Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics (Cardiology), Baylor College of Medicine, Houston, TX, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
41
|
Manca P, Nuzzi V, Cannatà A, Castrichini M, Bromage DI, De Luca A, Stolfo D, Schulz U, Merlo M, Sinagra G. The right ventricular involvement in dilated cardiomyopathy: prevalence and prognostic implications of the often-neglected child. Heart Fail Rev 2022; 27:1795-1805. [PMID: 35315505 PMCID: PMC9388461 DOI: 10.1007/s10741-022-10229-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/15/2022] [Indexed: 12/02/2022]
Abstract
Dilated cardiomyopathy (DCM) is a primary heart muscle disease characterized by left or biventricular systolic impairment. Historically, most of the clinical attention has been devoted to the evaluation of left ventricular function and morphology, while right ventricle (RV) has been for many years the forgotten chamber. Recently, progresses in cardiac imaging gave clinicians precious tools for the evaluation of RV, raising the awareness of the importance of biventricular assessment in DCM. Indeed, RV involvement is far from being uncommon in DCM, and the presence of right ventricular dysfunction (RVD) is one of the major negative prognostic determinants in DCM patients. However, some aspects such as the possible role of specific genetic mutations in determining the biventricular phenotype in DCM, or the lack of specific treatments able to primarily counteract RVD, still need research. In this review, we summarized the current knowledge on RV involvement in DCM, giving an overview on the epidemiology and pathogenetic mechanisms implicated in determining RVD. Furthermore, we discussed the imaging techniques to evaluate RV function and the role of RV failure in advanced heart failure.
Collapse
Affiliation(s)
- Paolo Manca
- Division of Cardiology, Cardiovascular Department, Azienda Sanitaria Universitaria Integrata Giuliana Isontina (ASUGI), University of Trieste, Via Valdoni 7, 34149, Trieste, Italy
| | - Vincenzo Nuzzi
- Division of Cardiology, Cardiovascular Department, Azienda Sanitaria Universitaria Integrata Giuliana Isontina (ASUGI), University of Trieste, Via Valdoni 7, 34149, Trieste, Italy
| | - Antonio Cannatà
- Division of Cardiology, Cardiovascular Department, Azienda Sanitaria Universitaria Integrata Giuliana Isontina (ASUGI), University of Trieste, Via Valdoni 7, 34149, Trieste, Italy.,Department of Cardiovascular Science, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Matteo Castrichini
- Division of Cardiology, Cardiovascular Department, Azienda Sanitaria Universitaria Integrata Giuliana Isontina (ASUGI), University of Trieste, Via Valdoni 7, 34149, Trieste, Italy
| | - Daniel I Bromage
- Department of Cardiovascular Science, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Antonio De Luca
- Division of Cardiology, Cardiovascular Department, Azienda Sanitaria Universitaria Integrata Giuliana Isontina (ASUGI), University of Trieste, Via Valdoni 7, 34149, Trieste, Italy
| | - Davide Stolfo
- Division of Cardiology, Cardiovascular Department, Azienda Sanitaria Universitaria Integrata Giuliana Isontina (ASUGI), University of Trieste, Via Valdoni 7, 34149, Trieste, Italy.,Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Uwe Schulz
- Department of Cardiac Surgery, Heart Center, University of Leipzig, Leipzig, Germany
| | - Marco Merlo
- Division of Cardiology, Cardiovascular Department, Azienda Sanitaria Universitaria Integrata Giuliana Isontina (ASUGI), University of Trieste, Via Valdoni 7, 34149, Trieste, Italy.
| | - Gianfranco Sinagra
- Division of Cardiology, Cardiovascular Department, Azienda Sanitaria Universitaria Integrata Giuliana Isontina (ASUGI), University of Trieste, Via Valdoni 7, 34149, Trieste, Italy
| |
Collapse
|
42
|
Antisense Therapy Attenuates Phospholamban p.(Arg14del) Cardiomyopathy in Mice and Reverses Protein Aggregation. Int J Mol Sci 2022; 23:ijms23052427. [PMID: 35269571 PMCID: PMC8909937 DOI: 10.3390/ijms23052427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/16/2022] [Accepted: 02/21/2022] [Indexed: 12/26/2022] Open
Abstract
Inherited cardiomyopathy caused by the p.(Arg14del) pathogenic variant of the phospholamban (PLN) gene is characterized by intracardiomyocyte PLN aggregation and can lead to severe dilated cardiomyopathy. We recently reported that pre-emptive depletion of PLN attenuated heart failure (HF) in several cardiomyopathy models. Here, we investigated if administration of a Pln-targeting antisense oligonucleotide (ASO) could halt or reverse disease progression in mice with advanced PLN-R14del cardiomyopathy. To this aim, homozygous PLN-R14del (PLN-R14 Δ/Δ) mice received PLN-ASO injections starting at 5 or 6 weeks of age, in the presence of moderate or severe HF, respectively. Mice were monitored for another 4 months with echocardiographic analyses at several timepoints, after which cardiac tissues were examined for pathological remodeling. We found that vehicle-treated PLN-R14 Δ/Δ mice continued to develop severe HF, and reached a humane endpoint at 8.1 ± 0.5 weeks of age. Both early and late PLN-ASO administration halted further cardiac remodeling and dysfunction shortly after treatment start, resulting in a life span extension to at least 22 weeks of age. Earlier treatment initiation halted disease development sooner, resulting in better heart function and less remodeling at the study endpoint. PLN-ASO treatment almost completely eliminated PLN aggregates, and normalized levels of autophagic proteins. In conclusion, these findings indicate that PLN-ASO therapy may have beneficial outcomes in PLN-R14del cardiomyopathy when administered after disease onset. Although existing tissue damage was not reversed, further cardiomyopathy progression was stopped, and PLN aggregates were resolved.
Collapse
|
43
|
de Boer RA, Heymans S, Backs J, Carrier L, Coats AJS, Dimmeler S, Eschenhagen T, Filippatos G, Gepstein L, Hulot JS, Knöll R, Kupatt C, Linke WA, Seidman CE, Tocchetti CG, van der Velden J, Walsh R, Seferovic PM, Thum T. Targeted therapies in genetic dilated and hypertrophic cardiomyopathies: From molecular mechanisms to therapeutic targets. Eur J Heart Fail 2021; 24:406-420. [PMID: 34969177 PMCID: PMC9305112 DOI: 10.1002/ejhf.2414] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 11/15/2022] Open
Abstract
Genetic cardiomyopathies are disorders of the cardiac muscle, most often explained by pathogenic mutations in genes encoding sarcomere, cytoskeleton, or ion channel proteins. Clinical phenotypes such as heart failure and arrhythmia are classically treated with generic drugs, but aetiology‐specific and targeted treatments are lacking. As a result, cardiomyopathies still present a major burden to society, and affect many young and older patients. The Translational Committee of the Heart Failure Association (HFA) and the Working Group of Myocardial Function of the European Society of Cardiology (ESC) organized a workshop to discuss recent advances in molecular and physiological studies of various forms of cardiomyopathies. The study of cardiomyopathies has intensified after several new study setups became available, such as induced pluripotent stem cells, three‐dimensional printing of cells, use of scaffolds and engineered heart tissue, with convincing human validation studies. Furthermore, our knowledge on the consequences of mutated proteins has deepened, with relevance for cellular homeostasis, protein quality control and toxicity, often specific to particular cardiomyopathies, with precise effects explaining the aberrations. This has opened up new avenues to treat cardiomyopathies, using contemporary techniques from the molecular toolbox, such as gene editing and repair using CRISPR‐Cas9 techniques, antisense therapies, novel designer drugs, and RNA therapies. In this article, we discuss the connection between biology and diverse clinical presentation, as well as promising new medications and therapeutic avenues, which may be instrumental to come to precision medicine of genetic cardiomyopathies.
Collapse
Affiliation(s)
- Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Stephane Heymans
- Department of Cardiology, Maastricht University Medical Center (MUMC+), PO Box 5800, 6202, AZ, Maastricht, the Netherlands.,Department of Cardiovascular Sciences, University of Leuven, Belgium
| | - Johannes Backs
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | | | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Gerasimos Filippatos
- Department of Cardiology, National and Kapodistrian University of Athens, School of Medicine, Attikon University Hospital, Athens, Greece
| | - Lior Gepstein
- Department of Cardiology, Rambam Health Care Campus, Haaliya Street, 31096, Haifa, Israel
| | - Jean-Sebastien Hulot
- Université de Paris, INSERM, PARCC, F-75006, Paris, France.,CIC1418 and DMU CARTE, AP- HP, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| | - Ralph Knöll
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Heart and Vascular Theme, Karolinska Institute, Stockholm, SE-171 77, Sweden.,Bioscience, Cardiovascular, Renal & Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christian Kupatt
- Department of Cardiology, University Clinic rechts der Isar, Technical University of Munich, Germany and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance
| | - Wolfgang A Linke
- Institute of Physiology II, University Hospital Muenster, Robert-Koch-Str. 27B, 48149, Muenster, Germany
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA.,Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Howard Hughes Medical Institute, Harvard University, Boston, MA, USA
| | - C Gabriele Tocchetti
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI); Interdepartmental Center for Clinical and Translational Research (CIRCET); Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Amsterdam Cardiovascular Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Roddy Walsh
- Department of Clinical and Experimental Cardiology, Amsterdam UMC, Amsterdam Cardiovascular Sciences, University of Amsterdam, Heart Center, Amsterdam, The Netherlands
| | - Petar M Seferovic
- Serbian Academy of Sciences and Arts, Belgrade, 11000, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, 11000, Serbia
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| |
Collapse
|
44
|
Buonaiuto G, Desideri F, Taliani V, Ballarino M. Muscle Regeneration and RNA: New Perspectives for Ancient Molecules. Cells 2021; 10:cells10102512. [PMID: 34685492 PMCID: PMC8533951 DOI: 10.3390/cells10102512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/21/2022] Open
Abstract
The ability of the ribonucleic acid (RNA) to self-replicate, combined with a unique cocktail of chemical properties, suggested the existence of an RNA world at the origin of life. Nowadays, this hypothesis is supported by innovative high-throughput and biochemical approaches, which definitively revealed the essential contribution of RNA-mediated mechanisms to the regulation of fundamental processes of life. With the recent development of SARS-CoV-2 mRNA-based vaccines, the potential of RNA as a therapeutic tool has received public attention. Due to its intrinsic single-stranded nature and the ease with which it is synthesized in vitro, RNA indeed represents the most suitable tool for the development of drugs encompassing every type of human pathology. The maximum effectiveness and biochemical versatility is achieved in the guise of non-coding RNAs (ncRNAs), which are emerging as multifaceted regulators of tissue specification and homeostasis. Here, we report examples of coding and ncRNAs involved in muscle regeneration and discuss their potential as therapeutic tools. Small ncRNAs, such as miRNA and siRNA, have been successfully applied in the treatment of several diseases. The use of longer molecules, such as lncRNA and circRNA, is less advanced. However, based on the peculiar properties discussed below, they represent an innovative pool of RNA biomarkers and possible targets of clinical value.
Collapse
MESH Headings
- Animals
- Biomarkers/metabolism
- COVID-19
- Homeostasis
- Humans
- Mice
- MicroRNAs/metabolism
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/virology
- Myocardium/metabolism
- Origin of Life
- RNA, Circular
- RNA, Long Noncoding/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- RNA, Small Untranslated/genetics
- RNA, Untranslated/genetics
- RNA, Viral/metabolism
- Regeneration
- SARS-CoV-2/genetics
Collapse
Affiliation(s)
- Giulia Buonaiuto
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (G.B.); (F.D.); (V.T.)
| | - Fabio Desideri
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (G.B.); (F.D.); (V.T.)
- Center for Life Nano & Neuro-Science of Instituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Valeria Taliani
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (G.B.); (F.D.); (V.T.)
| | - Monica Ballarino
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (G.B.); (F.D.); (V.T.)
- Correspondence:
| |
Collapse
|