1
|
Ragusa R, Caselli C. Focus on cardiac troponin complex: From gene expression to cardiomyopathy. Genes Dis 2024; 11:101263. [PMID: 39211905 PMCID: PMC11357864 DOI: 10.1016/j.gendis.2024.101263] [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: 10/03/2023] [Revised: 01/26/2024] [Accepted: 02/21/2024] [Indexed: 09/04/2024] Open
Abstract
The cardiac troponin complex (cTn) is a regulatory component of sarcomere. cTn consists of three subunits: cardiac troponin C (cTnC), which confers Ca2+ sensitivity to muscle; cTnI, which inhibits the interaction of cross-bridge of myosin with thin filament during diastole; and cTnT, which has multiple roles in sarcomere, such as promoting the link between the cTnI-cTnC complex and tropomyosin within the thin filament and influencing Ca2+ sensitivity of cTn and force development during contraction. Conditions that interfere with interactions within cTn and/or other thin filament proteins can be key factors in the regulation of cardiac contraction. These conditions include alterations in myofilament Ca2+ sensitivity, direct changes in cTn function, and triggering downstream events that lead to adverse cardiac remodeling and impairment of heart function. This review describes gene expression and post-translational modifications of cTn as well as the conditions that can adversely affect the delicate balance among the components of cTn, thereby promoting contractile dysfunction.
Collapse
Affiliation(s)
- Rosetta Ragusa
- Institute of Clinical Physiology, CNR, via Moruzzi 1, Pisa 56124, Italy
| | - Chiara Caselli
- Institute of Clinical Physiology, CNR, via Moruzzi 1, Pisa 56124, Italy
- Fondazione Toscana Gabriele Monasterio, via Moruzzi 1, Pisa 56124, Italy
| |
Collapse
|
2
|
Huang Y, Zhang Y, Wang Z, Miao L, Tan P, Guan Y, Ran Y, Feng X, Wang Y, Guo Y, Guo X. Modified mRNA-based gene editing reveals sarcomere-based regulation of gene expression in human induced-pluripotent stem cell-derived cardiomyocytes. Int Immunopharmacol 2024; 143:113378. [PMID: 39423657 DOI: 10.1016/j.intimp.2024.113378] [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: 07/18/2024] [Revised: 09/17/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
Mutations in genes coding sarcomere components are the major causes of human inherited cardiomyopathy. Genome editing is widely applied to genetic modification of human pluripotent stem cells (hPSCs) before hPSCs were differentiated into cardiomyocytes to model cardiomyopathy. Whether genetic mutations influence the early hPSC differentiation process or solely the terminally differentiated cardiomyocytes during cardiac pathogenesis remains challenging to distinguish. To solve this problem, here we harnessed chemically modified mRNA (modRNA) and synthetic single-guide RNA to develop an efficient genome editing approach in hPSC-derived cardiomyocytes (hPSC-CMs). We showed that modRNA-based CRISPR/Cas9 mutagenesis of TNNT2, the coding gene for cardiac troponin T, results in sarcomere disassembly and contractile dysfunction in hPSC-CMs. These structural and functional phenotypes were associated with profound downregulation of oxidative phosphorylation genes and upregulation of cardiac stress markers NPPA and NPPB. These data confirmed that sarcomeres regulate gene expression in hPSC-CMs and highlighted the RNA technology as a powerful tool to achieve stage-specific genome editing during hPSC differentiation.
Collapse
Affiliation(s)
- Yuqing Huang
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yueyang Zhang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China
| | - Ze Wang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China
| | - Lei Miao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Pingping Tan
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuting Guan
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuqing Ran
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xing Feng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yijia Wang
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuxuan Guo
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China.
| | - Xiaoling Guo
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
3
|
Yang Y, Rivera L, Fang S, Cavalier M, Suris A, Zhou Y, Huang Y. Maternal high-fat diet alters Tet-mediated epigenetic regulation during heart development. iScience 2024; 27:110631. [PMID: 39262804 PMCID: PMC11388159 DOI: 10.1016/j.isci.2024.110631] [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: 01/10/2024] [Revised: 05/24/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
Imbalanced dietary intake, such as a high-fat diet (HFD) during pregnancy, has been associated with adverse offspring outcomes. Metabolic stress from imbalanced food intake alters the function of epigenetic regulators, resulting in abnormal transcriptional outputs in embryos to cause congenital disorders. We report herein that maternal HFD exposure causes metabolic changes in pregnant mice and non-compaction cardiomyopathy (NCC) in E15.5 embryos, accompanied by decreased 5-hydroxymethylcytosine (5hmC) levels and altered chromatin accessibility in embryonic heart tissues. Remarkably, maternal vitamin C supplementation mitigates these detrimental effects, likely by restoring iron, a cofactor for Tet enzymes, in a reduced state. Using a genetic approach, we further demonstrated that the cardioprotective benefits of vitamin C under HFD conditions are attributable to enhanced Tet activity. Our results highlight an interaction between maternal diet, specifically HFD or vitamin C, and epigenetic modifications during early heart development, emphasizing the importance of balanced maternal nutrition for healthy embryonic development.
Collapse
Affiliation(s)
- Yuhan Yang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Logan Rivera
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Shaohai Fang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Maryn Cavalier
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Ashley Suris
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
- Department of Translational Medical Sciences, School of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Yun Huang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
- Department of Translational Medical Sciences, School of Medicine, Texas A&M University, Houston, TX 77030, USA
| |
Collapse
|
4
|
Kim OH, Kim J, Kim Y, Lee S, Lee BH, Kim BJ, Park HY, Park MH. Exploring novel MYH7 gene variants using in silico analyses in Korean patients with cardiomyopathy. BMC Med Genomics 2024; 17:225. [PMID: 39237976 PMCID: PMC11378590 DOI: 10.1186/s12920-024-02000-8] [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: 02/23/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Pathogenic variants of MYH7, which encodes the beta-myosin heavy chain protein, are major causes of dilated and hypertrophic cardiomyopathy. METHODS In this study, we used whole-genome sequencing data to identify MYH7 variants in 397 patients with various cardiomyopathy subtypes who were participating in the National Project of Bio Big Data pilot study in Korea. We also performed in silico analyses to predict the pathogenicity of the novel variants, comparing them to known pathogenic missense variants. RESULTS We identified 27 MYH7 variants in 41 unrelated patients with cardiomyopathy, consisting of 20 previously known pathogenic/likely pathogenic variants, 2 variants of uncertain significance, and 5 novel variants. Notably, the pathogenic variants predominantly clustered within the myosin motor domain of MYH7. We confirmed that the novel identified variants could be pathogenic, as indicated by high prediction scores in the in silico analyses, including SIFT, Mutation Assessor, PROVEAN, PolyPhen-2, CADD, REVEL, MetaLR, MetaRNN, and MetaSVM. Furthermore, we assessed their damaging effects on protein dynamics and stability using DynaMut2 and Missense3D tools. CONCLUSIONS Overall, our study identified the distribution of MYH7 variants among patients with cardiomyopathy in Korea, offering new insights for improved diagnosis by enriching the data on the pathogenicity of novel variants using in silico tools and evaluating the function and structural stability of the MYH7 protein.
Collapse
Affiliation(s)
- Oc-Hee Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, 28159, Republic of Korea
| | - Jihyun Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, 28159, Republic of Korea
| | - Youngjun Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, 28159, Republic of Korea
| | - Soyoung Lee
- Department of Pediatrics, Hallym University Sacred Heart Hospital, Anyang, 14068, Republic of Korea
| | - Beom Hee Lee
- Medical Genetics Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicines, Seoul, 05505, Republic of Korea
| | - Bong-Jo Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, 28159, Republic of Korea
| | - Hyun-Young Park
- National Institute of Health, Cheongju, 28159, Republic of Korea
| | - Mi-Hyun Park
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, 28159, Republic of Korea.
| |
Collapse
|
5
|
Malinow I, Fong DC, Miyamoto M, Badran S, Hong CC. Pediatric dilated cardiomyopathy: a review of current clinical approaches and pathogenesis. Front Pediatr 2024; 12:1404942. [PMID: 38966492 PMCID: PMC11223501 DOI: 10.3389/fped.2024.1404942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/05/2024] [Indexed: 07/06/2024] Open
Abstract
Pediatric dilated cardiomyopathy (DCM) is a rare, yet life-threatening cardiovascular condition characterized by systolic dysfunction with biventricular dilatation and reduced myocardial contractility. Therapeutic options are limited with nearly 40% of children undergoing heart transplant or death within 2 years of diagnosis. Pediatric patients are currently diagnosed based on correlating the clinical picture with echocardiographic findings. Patient age, etiology of disease, and parameters of cardiac function significantly impact prognosis. Treatments for pediatric DCM aim to ameliorate symptoms, reduce progression of disease, and prevent life-threatening arrhythmias. Many therapeutic agents with known efficacy in adults lack the same evidence in children. Unlike adult DCM, the pathogenesis of pediatric DCM is not well understood as approximately two thirds of cases are classified as idiopathic disease. Children experience unique gene expression changes and molecular pathway activation in response to DCM. Studies have pointed to a significant genetic component in pediatric DCM, with variants in genes related to sarcomere and cytoskeleton structure implicated. In this regard, pediatric DCM can be considered pediatric manifestations of inherited cardiomyopathy syndromes. Yet exciting recent studies in infantile DCM suggest that this subset has a distinct etiology involving defective postnatal cardiac maturation, such as the failure of programmed centrosome breakdown in cardiomyocytes. Improved knowledge of pathogenesis is central to developing child-specific treatment approaches. This review aims to discuss the established biological pathogenesis of pediatric DCM, current clinical guidelines, and promising therapeutic avenues, highlighting differences from adult disease. The overarching goal is to unravel the complexities surrounding this condition to facilitate the advancement of novel therapeutic interventions and improve prognosis and overall quality of life for pediatric patients affected by DCM.
Collapse
Affiliation(s)
- Ian Malinow
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Daniel C. Fong
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Matthew Miyamoto
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sarah Badran
- Department of Pediatric Cardiology, Michigan State University College of Human Medicine Helen Devos Children’s Hospital, Grand Rapids, MI, United States
| | - Charles C. Hong
- Department of Medicine, Division of Cardiology, Michigan State University College of Human Medicine, East Lansing, MI, United States
| |
Collapse
|
6
|
Greenberg L, Tom Stump W, Lin Z, Bredemeyer AL, Blackwell T, Han X, Greenberg AE, Garcia BA, Lavine KJ, Greenberg MJ. Harnessing molecular mechanism for precision medicine in dilated cardiomyopathy caused by a mutation in troponin T. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588306. [PMID: 38645235 PMCID: PMC11030379 DOI: 10.1101/2024.04.05.588306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Familial dilated cardiomyopathy (DCM) is frequently caused by autosomal dominant point mutations in genes involved in diverse cellular processes, including sarcomeric contraction. While patient studies have defined the genetic landscape of DCM, genetics are not currently used in patient care, and patients receive similar treatments regardless of the underlying mutation. It has been suggested that a precision medicine approach based on the molecular mechanism of the underlying mutation could improve outcomes; however, realizing this approach has been challenging due to difficulties linking genotype and phenotype and then leveraging this information to identify therapeutic approaches. Here, we used multiscale experimental and computational approaches to test whether knowledge of molecular mechanism could be harnessed to connect genotype, phenotype, and drug response for a DCM mutation in troponin T, deletion of K210. Previously, we showed that at the molecular scale, the mutation reduces thin filament activation. Here, we used computational modeling of this molecular defect to predict that the mutant will reduce cellular and tissue contractility, and we validated this prediction in human cardiomyocytes and engineered heart tissues. We then used our knowledge of molecular mechanism to computationally model the effects of a small molecule that can activate the thin filament. We demonstrate experimentally that the modeling correctly predicts that the small molecule can partially rescue systolic dysfunction at the expense of diastolic function. Taken together, our results demonstrate how molecular mechanism can be harnessed to connect genotype and phenotype and inspire strategies to optimize mechanism-based therapeutics for DCM.
Collapse
Affiliation(s)
- Lina Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - W. Tom Stump
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Zongtao Lin
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Andrea L. Bredemeyer
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Thomas Blackwell
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xian Han
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Akiva E. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Benjamin A. Garcia
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kory J. Lavine
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
7
|
Coscarella IL, Landim-Vieira M, Rastegarpouyani H, Chase PB, Irianto J, Pinto JR. Nucleus Mechanosensing in Cardiomyocytes. Int J Mol Sci 2023; 24:13341. [PMID: 37686151 PMCID: PMC10487505 DOI: 10.3390/ijms241713341] [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: 07/14/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Cardiac muscle contraction is distinct from the contraction of other muscle types. The heart continuously undergoes contraction-relaxation cycles throughout an animal's lifespan. It must respond to constantly varying physical and energetic burdens over the short term on a beat-to-beat basis and relies on different mechanisms over the long term. Muscle contractility is based on actin and myosin interactions that are regulated by cytoplasmic calcium ions. Genetic variants of sarcomeric proteins can lead to the pathophysiological development of cardiac dysfunction. The sarcomere is physically connected to other cytoskeletal components. Actin filaments, microtubules and desmin proteins are responsible for these interactions. Therefore, mechanical as well as biochemical signals from sarcomeric contractions are transmitted to and sensed by other parts of the cardiomyocyte, particularly the nucleus which can respond to these stimuli. Proteins anchored to the nuclear envelope display a broad response which remodels the structure of the nucleus. In this review, we examine the central aspects of mechanotransduction in the cardiomyocyte where the transmission of mechanical signals to the nucleus can result in changes in gene expression and nucleus morphology. The correlation of nucleus sensing and dysfunction of sarcomeric proteins may assist the understanding of a wide range of functional responses in the progress of cardiomyopathic diseases.
Collapse
Affiliation(s)
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Hosna Rastegarpouyani
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
- Institute for Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Prescott Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Jerome Irianto
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
8
|
Langa P, Shafaattalab S, Goldspink PH, Wolska BM, Fernandes AA, Tibbits GF, Solaro RJ. A perspective on Notch signalling in progression and arrhythmogenesis in familial hypertrophic and dilated cardiomyopathies. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220176. [PMID: 37122209 PMCID: PMC10150215 DOI: 10.1098/rstb.2022.0176] [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: 09/14/2022] [Accepted: 10/13/2022] [Indexed: 05/02/2023] Open
Abstract
In this perspective, we discussed emerging data indicating a role for Notch signalling in inherited disorders of the heart failure with focus on hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) linked to variants of genes encoding mutant proteins of the sarcomere. We recently reported an upregulation of elements in the Notch signalling cascade in cardiomyocytes derived from human inducible pluripotent stem cells expressing a TNNT2 variant encoding cardiac troponin T (cTnT-I79N+/-), which induces hypertrophy, remodelling, abnormalities in excitation-contraction coupling and electrical instabilities (Shafaattalab S et al. 2021 Front. Cell Dev. Biol. 9, 787581. (doi:10.3389/fcell.2021.787581)). Our search of the literature revealed the novelty of this finding and stimulated us to discuss potential connections between the Notch signalling pathway and familial cardiomyopathies. Our considerations focused on the potential role of these interactions in arrhythmias, microvascular ischaemia, and fibrosis. This finding underscored a need to consider the role of Notch signalling in familial cardiomyopathies which are trigged by sarcomere mutations engaging mechano-signalling pathways for which there is evidence of a role for Notch signalling with crosstalk with Hippo signalling. Our discussion included a role for both cardiac myocytes and non-cardiac myocytes in progression of HCM and DCM. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Paulina Langa
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| | - Sanam Shafaattalab
- Molecular Biology and Biochemistry; BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4; Simon Fraser University Burnaby, British Columbia, V5A 4H4, Canada
| | - Paul H. Goldspink
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| | - Beata M. Wolska
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
- Department of Medicine, Division of Cardiology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Aurelia A. Fernandes
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| | - Glen F. Tibbits
- Molecular Biology and Biochemistry; BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4; Simon Fraser University Burnaby, British Columbia, V5A 4H4, Canada
| | - R. John Solaro
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| |
Collapse
|
9
|
A Novel Nonsense Pathogenic TTN Variant Identified in a Patient with Severe Dilated Cardiomyopathy. Curr Issues Mol Biol 2023; 45:2422-2430. [PMID: 36975527 PMCID: PMC10047881 DOI: 10.3390/cimb45030157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Both genetic and environmental factors contribute to the development of dilated cardiomyopathy. Among the genes involved, TTN mutations, including truncated variants, explain 25% of DCM cases. We performed genetic counseling and analysis on a 57-year-old woman diagnosed with severe DCM and presenting relevant acquired risk factors for DCM (hypertension, diabetes, smoking habit, and/or previous alcohol and cocaine abuse) and with a family history of both DCM and sudden cardiac death. The left ventricular systolic function, as assessed by standard echocardiography, was 20%. The genetic analysis performed using TruSight Cardio panel, including 174 genes related to cardiac genetic diseases, revealed a novel nonsense TTN variant (TTN:c.103591A > T, p.Lys34531*), falling within the M-band region of the titin protein. This region is known for its important role in maintaining the structure of the sarcomere and in promoting sarcomerogenesis. The identified variant was classified as likely pathogenic based on ACMG criteria. The current results support the need of genetic analysis in the presence of a family history, even when relevant acquired risk factors for DCM may have contributed to the severity of the disease.
Collapse
|
10
|
Barrick SK, Garg A, Greenberg L, Zhang S, Lin CY, Stitziel NO, Greenberg MJ. Functional assays reveal the pathogenic mechanism of a de novo tropomyosin variant identified in patient with dilated cardiomyopathy. J Mol Cell Cardiol 2023; 176:58-67. [PMID: 36739943 PMCID: PMC11285302 DOI: 10.1016/j.yjmcc.2023.01.014] [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: 09/06/2022] [Revised: 01/09/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure and a major indicator for heart transplant. Human genetic studies have identified over a thousand causal mutations for DCM in genes involved in a variety of cellular processes, including sarcomeric contraction. A substantial clinical challenge is determining the pathogenicity of novel variants in disease-associated genes. This challenge of connecting genotype and phenotype has frustrated attempts to develop effective, mechanism-based treatments for patients. Here, we identified a de novo mutation (T237S) in TPM1, the gene that encodes the thin filament protein tropomyosin, in a patient with DCM and conducted in vitro experiments to characterize the pathogenicity of this novel variant. We expressed recombinant mutant protein, reconstituted it into thin filaments, and examined the effects of the mutation on thin filament function. We show that the mutation reduces the calcium sensitivity of thin filament activation, as previously seen for known pathogenic mutations. Mechanistically, this shift is due to mutation-induced changes in tropomyosin positioning along the thin filament. We demonstrate that the thin filament activator omecamtiv mecarbil restores the calcium sensitivity of thin filaments regulated by the mutant tropomyosin, which lays the foundation for additional experiments to explore the therapeutic potential of this drug for patients harboring the T237S mutation. Taken together, our results suggest that the TPM1 T237S mutation is likely pathogenic and demonstrate how functional in vitro characterization of pathogenic protein variants in the lab might guide precision medicine in the clinic.
Collapse
Affiliation(s)
- Samantha K Barrick
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ankit Garg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lina Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shanshan Zhang
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nathan O Stitziel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
11
|
Risi CM, Belknap B, White HD, Dryden K, Pinto JR, Chase PB, Galkin VE. High-resolution cryo-EM structure of the junction region of the native cardiac thin filament in relaxed state. PNAS NEXUS 2023; 2:pgac298. [PMID: 36712934 PMCID: PMC9832952 DOI: 10.1093/pnasnexus/pgac298] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Cardiac contraction depends on molecular interactions among sarcomeric proteins coordinated by the rising and falling intracellular Ca2+ levels. Cardiac thin filament (cTF) consists of two strands composed of actin, tropomyosin (Tm), and equally spaced troponin (Tn) complexes forming regulatory units. Tn binds Ca2+ to move Tm strand away from myosin-binding sites on actin to enable actomyosin cross-bridges required for force generation. The Tn complex has three subunits-Ca2+-binding TnC, inhibitory TnI, and Tm-binding TnT. Tm strand is comprised of adjacent Tm molecules that overlap "head-to-tail" along the actin filament. The N-terminus of TnT (e.g., TnT1) binds to the Tm overlap region to form the cTF junction region-the region that connects adjacent regulatory units and confers to cTF internal cooperativity. Numerous studies have predicted interactions among actin, Tm, and TnT1 within the junction region, although a direct structural description of the cTF junction region awaited completion. Here, we report a 3.8 Å resolution cryo-EM structure of the native cTF junction region at relaxing (pCa 8) Ca2+ conditions. We provide novel insights into the "head-to-tail" interactions between adjacent Tm molecules and interactions between the Tm junction with F-actin. We demonstrate how TnT1 stabilizes the Tm overlap region via its interactions with the Tm C- and N-termini and actin. Our data show that TnT1 works as a joint that anchors the Tm overlap region to actin, which stabilizes the relaxed state of the cTF. Our structure provides insight into the molecular basis of cardiac diseases caused by missense mutations in TnT1.
Collapse
Affiliation(s)
- Cristina M Risi
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Betty Belknap
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Howard D White
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Kelly Dryden
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22904, USA
| | - Jose R Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32304, USA
| | - P Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Vitold E Galkin
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| |
Collapse
|
12
|
Pham JH, Giudicessi JR, Tweet MS, Boucher L, Newman DB, Geske JB. Tale of two hearts: a TNNT2 hypertrophic cardiomyopathy case report. Front Cardiovasc Med 2023; 10:1167256. [PMID: 37180798 PMCID: PMC10174446 DOI: 10.3389/fcvm.2023.1167256] [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: 02/16/2023] [Accepted: 04/07/2023] [Indexed: 05/16/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a heritable cardiomyopathy that is predominantly caused by pathogenic mutations in sarcomeric proteins. Here we report two individuals, a mother and her daughter, both heterozygous carriers of the same HCM-causing mutation in cardiac Troponin T (TNNT2). Despite sharing an identical pathogenic variant, the two individuals had very different manifestations of the disease. While one patient presented with sudden cardiac death, recurrent tachyarrhythmia, and findings of massive left ventricular hypertrophy, the other patient manifested with extensive abnormal myocardial delayed enhancement despite normal ventricular wall thickness and has remained relatively asymptomatic. Recognition of the marked incomplete penetrance and variable expressivity possible in a single TNNT2-positive family has potential to guide HCM patient care.
Collapse
Affiliation(s)
- Justin H. Pham
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN, United States
| | - John R. Giudicessi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester MN, United States
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Marysia S. Tweet
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester MN, United States
| | - Lauren Boucher
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester MN, United States
| | - D. Brian Newman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester MN, United States
| | - Jeffrey B. Geske
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester MN, United States
- Correspondence: Jeffrey B. Geske
| |
Collapse
|
13
|
Perea-Gil I, Seeger T, Bruyneel AAN, Termglinchan V, Monte E, Lim EW, Vadgama N, Furihata T, Gavidia AA, Arthur Ataam J, Bharucha N, Martinez-Amador N, Ameen M, Nair P, Serrano R, Kaur B, Feyen DAM, Diecke S, Snyder MP, Metallo CM, Mercola M, Karakikes I. Serine biosynthesis as a novel therapeutic target for dilated cardiomyopathy. Eur Heart J 2022; 43:3477-3489. [PMID: 35728000 PMCID: PMC9794189 DOI: 10.1093/eurheartj/ehac305] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 04/14/2022] [Accepted: 05/24/2022] [Indexed: 12/30/2022] Open
Abstract
AIMS Genetic dilated cardiomyopathy (DCM) is a leading cause of heart failure. Despite significant progress in understanding the genetic aetiologies of DCM, the molecular mechanisms underlying the pathogenesis of familial DCM remain unknown, translating to a lack of disease-specific therapies. The discovery of novel targets for the treatment of DCM was sought using phenotypic sceening assays in induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) that recapitulate the disease phenotypes in vitro. METHODS AND RESULTS Using patient-specific iPSCs carrying a pathogenic TNNT2 gene mutation (p.R183W) and CRISPR-based genome editing, a faithful DCM model in vitro was developed. An unbiased phenotypic screening in TNNT2 mutant iPSC-derived cardiomyocytes (iPSC-CMs) with small molecule kinase inhibitors (SMKIs) was performed to identify novel therapeutic targets. Two SMKIs, Gö 6976 and SB 203580, were discovered whose combinatorial treatment rescued contractile dysfunction in DCM iPSC-CMs carrying gene mutations of various ontologies (TNNT2, TTN, LMNA, PLN, TPM1, LAMA2). The combinatorial SMKI treatment upregulated the expression of genes that encode serine, glycine, and one-carbon metabolism enzymes and significantly increased the intracellular levels of glucose-derived serine and glycine in DCM iPSC-CMs. Furthermore, the treatment rescued the mitochondrial respiration defects and increased the levels of the tricarboxylic acid cycle metabolites and ATP in DCM iPSC-CMs. Finally, the rescue of the DCM phenotypes was mediated by the activating transcription factor 4 (ATF4) and its downstream effector genes, phosphoglycerate dehydrogenase (PHGDH), which encodes a critical enzyme of the serine biosynthesis pathway, and Tribbles 3 (TRIB3), a pseudokinase with pleiotropic cellular functions. CONCLUSIONS A phenotypic screening platform using DCM iPSC-CMs was established for therapeutic target discovery. A combination of SMKIs ameliorated contractile and metabolic dysfunction in DCM iPSC-CMs mediated via the ATF4-dependent serine biosynthesis pathway. Together, these findings suggest that modulation of serine biosynthesis signalling may represent a novel genotype-agnostic therapeutic strategy for genetic DCM.
Collapse
Affiliation(s)
- Isaac Perea-Gil
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Timon Seeger
- Department of Medicine III, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Arne A N Bruyneel
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Vittavat Termglinchan
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Emma Monte
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Esther W Lim
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Nirmal Vadgama
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
| | - Takaaki Furihata
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexandra A Gavidia
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
| | - Jennifer Arthur Ataam
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Nike Bharucha
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Noel Martinez-Amador
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
| | - Mohamed Ameen
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Pooja Nair
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
| | - Ricardo Serrano
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Balpreet Kaur
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
| | - Dries A M Feyen
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sebastian Diecke
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mark Mercola
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ioannis Karakikes
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
14
|
Keyt LK, Duran JM, Bui QM, Chen C, Miyamoto MI, Silva Enciso J, Tardiff JC, Adler ED. Thin filament cardiomyopathies: A review of genetics, disease mechanisms, and emerging therapeutics. Front Cardiovasc Med 2022; 9:972301. [PMID: 36158814 PMCID: PMC9489950 DOI: 10.3389/fcvm.2022.972301] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022] Open
Abstract
All muscle contraction occurs due to the cyclical interaction between sarcomeric thin and thick filament proteins within the myocyte. The thin filament consists of the proteins actin, tropomyosin, Troponin C, Troponin I, and Troponin T. Mutations in these proteins can result in various forms of cardiomyopathy, including hypertrophic, restrictive, and dilated phenotypes and account for as many as 30% of all cases of inherited cardiomyopathy. There is significant evidence that thin filament mutations contribute to dysregulation of Ca2+ within the sarcomere and may have a distinct pathomechanism of disease from cardiomyopathy associated with thick filament mutations. A number of distinct clinical findings appear to be correlated with thin-filament mutations: greater degrees of restrictive cardiomyopathy and relatively less left ventricular (LV) hypertrophy and LV outflow tract obstruction than that seen with thick filament mutations, increased morbidity associated with heart failure, increased arrhythmia burden and potentially higher mortality. Most therapies that improve outcomes in heart failure blunt the neurohormonal pathways involved in cardiac remodeling, while most therapies for hypertrophic cardiomyopathy involve use of negative inotropes to reduce LV hypertrophy or septal reduction therapies to reduce LV outflow tract obstruction. None of these therapies directly address the underlying sarcomeric dysfunction associated with thin-filament mutations. With mounting evidence that thin filament cardiomyopathies occur through a distinct mechanism, there is need for therapies targeting the unique, underlying mechanisms tailored for each patient depending on a given mutation.
Collapse
Affiliation(s)
- Lucas K. Keyt
- Department of Internal Medicine, University of California, San Diego, San Diego, CA, United States
| | - Jason M. Duran
- Department of Cardiology, University of California, San Diego, San Diego, CA, United States
| | - Quan M. Bui
- Department of Cardiology, University of California, San Diego, San Diego, CA, United States
| | - Chao Chen
- Department of Cardiology, University of California, San Diego, San Diego, CA, United States
| | | | - Jorge Silva Enciso
- Department of Cardiology, University of California, San Diego, San Diego, CA, United States
| | - Jil C. Tardiff
- Department of Medicine and Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | - Eric D. Adler
- Department of Cardiology, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
15
|
Coscarella IL, Landim-Vieira M, Pinto JR, Chelko SP. Arrhythmogenic Cardiomyopathy: Exercise Pitfalls, Role of Connexin-43, and Moving beyond Antiarrhythmics. Int J Mol Sci 2022; 23:ijms23158753. [PMID: 35955883 PMCID: PMC9369094 DOI: 10.3390/ijms23158753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 12/11/2022] Open
Abstract
Arrhythmogenic Cardiomyopathy (ACM), a Mendelian disorder that can affect both left and right ventricles, is most often associated with pathogenic desmosomal variants that can lead to fibrofatty replacement of the myocardium, a pathological hallmark of this disease. Current therapies are aimed to prevent the worsening of disease phenotypes and sudden cardiac death (SCD). Despite the use of implantable cardioverter defibrillators (ICDs) there is no present therapy that would mitigate the loss in electrical signal and propagation by these fibrofatty barriers. Recent studies have shown the influence of forced vs. voluntary exercise in a variety of healthy and diseased mice; more specifically, that exercised mice show increased Connexin-43 (Cx43) expression levels. Fascinatingly, increased Cx43 expression ameliorated the abnormal electrical signal conduction in the myocardium of diseased mice. These findings point to a major translational pitfall in current therapeutics for ACM patients, who are advised to completely cease exercising and already demonstrate reduced Cx43 levels at the myocyte intercalated disc. Considering cardiac dysfunction in ACM arises from the loss of cardiomyocytes and electrical signal conduction abnormalities, an increase in Cx43 expression-promoted by low to moderate intensity exercise and/or gene therapy-could very well improve cardiac function in ACM patients.
Collapse
Affiliation(s)
- Isabella Leite Coscarella
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32303, USA
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32303, USA
| | - José Renato Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32303, USA
| | - Stephen P. Chelko
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32303, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21215, USA
- Correspondence: ; Tel.: +1-850-644-2215
| |
Collapse
|
16
|
Migliore L, Galvagni F, Pierantozzi E, Sorrentino V, Rossi D. Allele-specific silencing by RNAi of R92Q and R173W mutations in cardiac troponin T. Exp Biol Med (Maywood) 2022; 247:805-814. [PMID: 35067102 PMCID: PMC9160939 DOI: 10.1177/15353702211072453] [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: 08/25/2021] [Accepted: 12/17/2021] [Indexed: 08/30/2024] Open
Abstract
Autosomal dominant mutations in sarcomere proteins such as the cardiac troponin T (TNNT2) are the main genetic causes of human hypertrophic cardiomyopathy and dilated cardiomyopathy. Allele-specific silencing by RNA interference (ASP-RNAi) holds promise as a therapeutic strategy for downregulating a single mutant allele with minimal suppression of the corresponding wild-type allele. Here, we propose ASP-RNAi as a possible strategy to specifically knockdown mutant alleles coding for R92Q and R173W mutant TNNT2 proteins, identified in hypertrophic and dilated cardiomyopathy, respectively. Different siRNAs were designed and validated by luciferase reporter assay and following analysis in HEK293T cells expressing either the wild-type or mutant TNNT2 alleles. This study is the first exploration of ASP-RNAi on TNNT2-R173W and TNNT2-R92Q mutations in vitro and gives a base for further application of allele silencing as a therapeutic treatment for TNNT2-mutation-associated cardiomyopathies.
Collapse
Affiliation(s)
- Loredana Migliore
- Department of Molecular and
Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Federico Galvagni
- Department of Biotechnology,
Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Enrico Pierantozzi
- Department of Molecular and
Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Vincenzo Sorrentino
- Department of Molecular and
Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Daniela Rossi
- Department of Molecular and
Developmental Medicine, University of Siena, 53100 Siena, Italy
| |
Collapse
|
17
|
Halas M, Langa P, Warren CM, Goldspink PH, Wolska BM, Solaro RJ. Effects of Sarcomere Activators and Inhibitors Targeting Myosin Cross-Bridges on Ca2+-Activation of Mature and Immature Mouse Cardiac Myofilaments. Mol Pharmacol 2022; 101:286-299. [PMID: 35236770 PMCID: PMC9092471 DOI: 10.1124/molpharm.121.000420] [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: 09/28/2021] [Accepted: 02/16/2022] [Indexed: 11/22/2022] Open
Abstract
We tested the hypothesis that isoform shifts in sarcomeres of the immature heart modify the effect of cardiac myosin-directed sarcomere inhibitors and activators. Omecamtiv mecarbil (OM) activates tension and is in clinical trials for the treatment of adult acute and chronic heart failure. Mavacamten (Mava) inhibits tension and is in clinical trials to relieve hyper-contractility and outflow obstruction in advanced genetic hypertrophic cardiomyopathy (HCM) linked commonly to mutations in sarcomeric proteins. To address the effect of these agents in developing sarcomeres we isolated heart fiber bundles, extracted membranes with Triton X-100, and measured tension developed over a range of Ca2+ concentrations with and without OM or Mava treatment. We made measurements in fiber bundles from hearts of adult non-transgenic controls (NTG) expressing cardiac troponin I (cTnI), and from hearts of transgenic mice (TG-ssTnI) expressing the fetal/neonatal form, slow skeletal troponin I (ssTnI). We also compared fibers from 7+14-day-old NTG mice expressing ssTnI and cTnI. These studies were repeated with 7+14-day old transgenic mice (TG-cTnT-R92Q) expressing a mutant form of cardiac TnT (cTnT) linked to HCM. OM increased Ca2+-sensitivity and decreased cooperative activation in both ssTnI- and cTnI- regulated myofilaments with a similar effect reducing sub-maximal tension in immature and mature myofilaments. Although Mava decreased tension similarly in cTnI- and ssTnI-regulated myofilaments controlled either by cTnT or cTnT-R92Q, its effect involved a depressed Ca2+-sensitivity in the mature cTnT-R92-myofilaments. Our data demonstrate an influence of myosin and thin filament-associated proteins on the actions of myosin-directed agents such as OM and Mava. Significance Statement The effects of myosin-targeted activators and inhibitors on Ca2+-activated tension in developing cardiac sarcomeres presented here provide novel, ex-vivo evidence as to their actions in early-stage cardiac disorders. These studies advance understanding of the molecular mechanisms of these agents that is important in pre-clinical studies employing sarcomere Ca2+-response as a screening approach. The data also inform the use of commonly immature cardiac myocytes generated from human inducible pluripotent stem cells in screening for sarcomere activators and inhibitors.
Collapse
Affiliation(s)
- Monika Halas
- Physiology and Biophysics, University of Illinois at Chicago, United States
| | - Paulina Langa
- Physiology and Biophysics, University of Illinois at Chicago, United States
| | - Chad M Warren
- Physiology and Biophysics, University of Illinois at Chicago, United States
| | - Paul H Goldspink
- Physiology and Biophysics, University of Illinois at Chicago, United States
| | - Beata M Wolska
- Department of Medicine, University of Illinois at Chicago, United States
| | - R John Solaro
- Physiology and Biophysics, University of Illinois at Chicago, United States
| |
Collapse
|
18
|
Proteomic analysis reveals rattlesnake venom modulation of proteins associated with cardiac tissue damage in mouse hearts. J Proteomics 2022; 258:104530. [PMID: 35182786 PMCID: PMC9308947 DOI: 10.1016/j.jprot.2022.104530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/19/2022] [Accepted: 02/13/2022] [Indexed: 11/20/2022]
Abstract
Snake envenomation is a common but neglected disease that affects millions of people around the world annually. Among venomous snake species in Brazil, the tropical rattlesnake (Crotalus durissus terrificus) accounts for the highest number of fatal envenomations and is responsible for the second highest number of bites. Snake venoms are complex secretions which, upon injection, trigger diverse physiological effects that can cause significant injury or death. The components of C. d. terrificus venom exhibit neurotoxic, myotoxic, hemotoxic, nephrotoxic, and cardiotoxic properties which present clinically as alteration of central nervous system function, motor paralysis, seizures, eyelid ptosis, ophthalmoplesia, blurred vision, coagulation disorders, rhabdomyolysis, myoglobinuria, and cardiorespiratory arrest. In this study, we focused on proteomic characterization of the cardiotoxic effects of C. d. terrificus venom in mouse models. We injected venom at half the lethal dose (LD50) into the gastrocnemius muscle. Mouse hearts were removed at set time points after venom injection (1 h, 6 h, 12 h, or 24 h) and subjected to trypsin digestion prior to high-resolution mass spectrometry. We analyzed the proteomic profiles of >1300 proteins and observed that several proteins showed noteworthy changes in their quantitative profiles, likely reflecting the toxic activity of venom components. Among the affected proteins were several associated with cellular deregulation and tissue damage. Changes in heart protein abundance offer insights into how they may work synergistically upon envenomation. SIGNIFICANCE: Venom of the tropical rattlesnake (Crotalus durissus terririficus) is known to be neurotoxic, myotoxic, nephrotoxic and cardiotoxic. Although there are several studies describing the biochemical effects of this venom, no work has yet described its proteomic effects in the cardiac tissue of mice. In this work, we describe the changes in several mouse cardiac proteins upon venom treatment. Our data shed new light on the clinical outcome of the envenomation by C. d. terrificus, as well as candidate proteins that could be investigated in efforts to improve current treatment approaches or in the development of novel therapeutic interventions in order to reduce mortality and morbidity resulting from envenomation.
Collapse
|
19
|
Kaviarasan V, Mohammed V, Veerabathiran R. Genetic predisposition study of heart failure and its association with cardiomyopathy. Egypt Heart J 2022; 74:5. [PMID: 35061126 PMCID: PMC8782994 DOI: 10.1186/s43044-022-00240-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF) is a clinical condition distinguished by structural and functional defects in the myocardium, which genetic and environmental factors can induce. HF is caused by various genetic factors that are both heterogeneous and complex. The incidence of HF varies depending on the definition and area, but it is calculated to be between 1 and 2% in developed countries. There are several factors associated with the progression of HF, ranging from coronary artery disease to hypertension, of which observed the most common genetic cause to be cardiomyopathy. The main objective of this study is to investigate heart failure and its association with cardiomyopathy with their genetic variants. The selected novel genes that have been linked to human inherited cardiomyopathy play a critical role in the pathogenesis and progression of HF. Research sources collected from the human gene mutation and several databases revealed that numerous genes are linked to cardiomyopathy and thus explained the hereditary influence of such a condition. Our findings support the understanding of the genetics aspect of HF and will provide more accurate evidence of the role of changing disease accuracy. Furthermore, a better knowledge of the molecular pathophysiology of genetically caused HF could contribute to the emergence of personalized therapeutics in future.
Collapse
Affiliation(s)
- Vaishak Kaviarasan
- Human Cytogenetics and Genomics Laboratory, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamilnadu, 603103, India
| | - Vajagathali Mohammed
- Human Cytogenetics and Genomics Laboratory, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamilnadu, 603103, India
| | - Ramakrishnan Veerabathiran
- Human Cytogenetics and Genomics Laboratory, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamilnadu, 603103, India.
| |
Collapse
|
20
|
Assum I, Krause J, Scheinhardt MO, Müller C, Hammer E, Börschel CS, Völker U, Conradi L, Geelhoed B, Zeller T, Schnabel RB, Heinig M. Tissue-specific multi-omics analysis of atrial fibrillation. Nat Commun 2022; 13:441. [PMID: 35064145 PMCID: PMC8782899 DOI: 10.1038/s41467-022-27953-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
Genome-wide association studies (GWAS) for atrial fibrillation (AF) have uncovered numerous disease-associated variants. Their underlying molecular mechanisms, especially consequences for mRNA and protein expression remain largely elusive. Thus, refined multi-omics approaches are needed for deciphering the underlying molecular networks. Here, we integrate genomics, transcriptomics, and proteomics of human atrial tissue in a cross-sectional study to identify widespread effects of genetic variants on both transcript (cis-eQTL) and protein (cis-pQTL) abundance. We further establish a novel targeted trans-QTL approach based on polygenic risk scores to determine candidates for AF core genes. Using this approach, we identify two trans-eQTLs and five trans-pQTLs for AF GWAS hits, and elucidate the role of the transcription factor NKX2-5 as a link between the GWAS SNP rs9481842 and AF. Altogether, we present an integrative multi-omics method to uncover trans-acting networks in small datasets and provide a rich resource of atrial tissue-specific regulatory variants for transcript and protein levels for cardiovascular disease gene prioritization.
Collapse
Affiliation(s)
- Ines Assum
- Computational Health Center, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), München, Germany
- Department of Informatics, Technical University Munich, München, Germany
| | - Julia Krause
- University Center of Cardiovascular Science, University Heart and Vascular Center Hamburg, Hamburg, Germany
- Partner site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany
| | - Markus O Scheinhardt
- Institute of Medical Biometry and Statistics, University of Lübeck, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Christian Müller
- University Center of Cardiovascular Science, University Heart and Vascular Center Hamburg, Hamburg, Germany
- Partner site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- Partner site Greifswald, DZHK (German Center for Cardiovascular Research), Greifswald, Germany
| | - Christin S Börschel
- Partner site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- Partner site Greifswald, DZHK (German Center for Cardiovascular Research), Greifswald, Germany
| | - Lenard Conradi
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Bastiaan Geelhoed
- Partner site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Tanja Zeller
- University Center of Cardiovascular Science, University Heart and Vascular Center Hamburg, Hamburg, Germany
- Partner site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany
| | - Renate B Schnabel
- Partner site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany.
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany.
| | - Matthias Heinig
- Computational Health Center, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), München, Germany.
- Department of Informatics, Technical University Munich, München, Germany.
- Partner site Munich, DZHK (German Center for Cardiovascular Research), Munich, Germany.
| |
Collapse
|
21
|
Barrick SK, Greenberg L, Greenberg MJ. A troponin T variant linked with pediatric dilated cardiomyopathy reduces the coupling of thin filament activation to myosin and calcium binding. Mol Biol Cell 2021; 32:1677-1689. [PMID: 34161147 PMCID: PMC8684737 DOI: 10.1091/mbc.e21-02-0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a significant cause of pediatric heart failure. Mutations in proteins that regulate cardiac muscle contraction can cause DCM; however, the mechanisms by which molecular-level mutations contribute to cellular dysfunction are not well understood. Better understanding of these mechanisms might enable the development of targeted therapeutics that benefit patient subpopulations with mutations that cause common biophysical defects. We examined the molecular- and cellular-level impacts of a troponin T variant associated with pediatric-onset DCM, R134G. The R134G variant decreased calcium sensitivity in an in vitro motility assay. Using stopped-flow and steady-state fluorescence measurements, we determined the molecular mechanism of the altered calcium sensitivity: R134G decouples calcium binding by troponin from the closed-to-open transition of the thin filament and decreases the cooperativity of myosin binding to regulated thin filaments. Consistent with the prediction that these effects would cause reduced force per sarcomere, cardiomyocytes carrying the R134G mutation are hypocontractile. They also show hallmarks of DCM that lie downstream of the initial insult, including disorganized sarcomeres and cellular hypertrophy. These results reinforce the importance of multiscale studies to fully understand mechanisms underlying human disease and highlight the value of mechanism-based precision medicine approaches for DCM.
Collapse
Affiliation(s)
- Samantha K Barrick
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Lina Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
22
|
Troponin T Mutation as a Cause of Left Ventricular Systolic Dysfunction in a Young Patient with Previous Surgical Correction of Aortic Coarctation. Biomolecules 2021; 11:biom11050696. [PMID: 34066613 PMCID: PMC8148585 DOI: 10.3390/biom11050696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/01/2021] [Accepted: 05/01/2021] [Indexed: 12/02/2022] Open
Abstract
Coarctation of the aorta is a leading cause of morbidity and mortality among adults with congenital heart disease (ACHD). Lifelong surveillance is mandatory to screen for possible long-term cardiovascular events. Left ventricular systolic dysfunction has been reported in association with recoarctation, and association with dilated cardiomyopathy (DCMP) is very rare. Herein, we report the case of a 19-year-old boy with coarctation of the aorta who complained of mild exertional dyspnea. Cardiac magnetic resonance revealed a moderately dilated, hypokinetic left ventricle (LV), with mildly reduced EF (45%), and residual isthmic coarctation was excluded. Genetic tests revealed a heterozygous missense variant in TNNT2 (NM_001001430.2): c.518G>A (p. Arg173Gln). This case highlights the role of careful history taking: a family history of cardiomyopathy should not be overlooked even when the clinical setting seems to suggest a predisposition to hemodynamic factors for LVSD.
Collapse
|
23
|
Li M, Xia S, Xu L, Tan H, Yang J, Wu Z, He X, Li L. Genetic analysis using targeted next-generation sequencing of sporadic Chinese patients with idiopathic dilated cardiomyopathy. J Transl Med 2021; 19:189. [PMID: 33941202 PMCID: PMC8091742 DOI: 10.1186/s12967-021-02832-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 04/13/2021] [Indexed: 11/10/2022] Open
Abstract
Background Inherited dilated cardiomyopathy (DCM) contributes to approximately 25% of idiopathic DCM cases, and the proportion is even higher in familial DCM patients. Most studies have focused on familial DCM, whereas the genetic profile of sporadic DCM in Chinese patients remains unknown. Methods Between June 2018 and September 2019, 24 patients diagnosed with idiopathic DCM without a family history were included in the present study. All patients underwent genetic screening for 80 DCM-related genes using targeted next-generation sequencing. Results By in silico analysis, 10 of 99 detected variants were considered pathogenic or likely-pathogenic, including seven TTN truncating variants (TTNtv), one in-frame deletion in TNNT2, one missense mutation in RBM20, and one frameshift deletion variant in FLNC. Of these variants, eight are reported for the first time. Conclusions Using targeted next-generation sequencing, potential genetic causes of idiopathic DCM were identified. Sarcomere mutations remained the most common genetic cause of inherited DCM in this cohort of sporadic Chinese DCM. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02832-3.
Collapse
Affiliation(s)
- Mingmin Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shuang Xia
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lan Xu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hong Tan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Junqing Yang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zejia Wu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xuyu He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Liwen Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
24
|
Gacita AM, Fullenkamp DE, Ohiri J, Pottinger T, Puckelwartz MJ, Nobrega MA, McNally EM. Genetic Variation in Enhancers Modifies Cardiomyopathy Gene Expression and Progression. Circulation 2021; 143:1302-1316. [PMID: 33478249 PMCID: PMC8009836 DOI: 10.1161/circulationaha.120.050432] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Inherited cardiomyopathy associates with a range of phenotypes, mediated by genetic and nongenetic factors. Noninherited cardiomyopathy also displays varying progression and outcomes. Expression of cardiomyopathy genes is under the regulatory control of promoters and enhancers, and human genetic variation in promoters and enhancers may contribute to this variability. METHODS We superimposed epigenomic profiling from hearts and cardiomyocytes, including promoter-capture chromatin conformation information, to identify enhancers for 2 cardiomyopathy genes, MYH7 and LMNA. Enhancer function was validated in human cardiomyocytes derived from induced pluripotent stem cells. We also conducted a genome-wide search to ascertain genomic variation in enhancers positioned to alter cardiac expression and correlated one of these variants to cardiomyopathy progression using biobank data. RESULTS Multiple enhancers were identified and validated for LMNA and MYH7, including a key enhancer that regulates the switch from MYH6 expression to MYH7 expression. Deletion of this enhancer resulted in a dose-dependent increase in MYH6 and faster contractile rate in engineered heart tissues. We searched for genomic variation in enhancer sequences across the genome, with a focus on nucleotide changes that create or interrupt transcription factor binding sites. The sequence variant, rs875908, disrupts a T-Box Transcription Factor 5 binding motif and maps to an enhancer region 2 kilobases from the transcriptional start site of MYH7. Gene editing to remove the enhancer that harbors this variant markedly reduced MYH7 expression in human cardiomyocytes. Using biobank-derived data, rs875908 associated with longitudinal echocardiographic features of cardiomyopathy. CONCLUSIONS Enhancers regulate cardiomyopathy gene expression, and genomic variation within these enhancer regions associates with cardiomyopathic progression over time. This integrated approach identified noncoding modifiers of cardiomyopathy and is applicable to other cardiac genes.
Collapse
Affiliation(s)
- Anthony M. Gacita
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Dominic E. Fullenkamp
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Joyce Ohiri
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Tess Pottinger
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Megan J. Puckelwartz
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | | | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| |
Collapse
|
25
|
Pettinato AM, Ladha FA, Mellert DJ, Legere N, Cohn R, Romano R, Thakar K, Chen YS, Hinson JT. Development of a Cardiac Sarcomere Functional Genomics Platform to Enable Scalable Interrogation of Human TNNT2 Variants. Circulation 2020; 142:2262-2275. [PMID: 33025817 DOI: 10.1161/circulationaha.120.047999] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Pathogenic TNNT2 variants are a cause of hypertrophic and dilated cardiomyopathies, which promote heart failure by incompletely understood mechanisms. The precise functional significance for 87% of TNNT2 variants remains undetermined, in part, because of a lack of functional genomics studies. The knowledge of which and how TNNT2 variants cause hypertrophic and dilated cardiomyopathies could improve heart failure risk determination, treatment efficacy, and therapeutic discovery, and provide new insights into cardiomyopathy pathogenesis, as well. METHODS We created a toolkit of human induced pluripotent stem cell models and functional assays using CRISPR/Cas9 to study TNNT2 variant pathogenicity and pathophysiology. Using human induced pluripotent stem cell-derived cardiomyocytes in cardiac microtissue and single-cell assays, we functionally interrogated 51 TNNT2 variants, including 30 pathogenic/likely pathogenic variants and 21 variants of uncertain significance. We used RNA sequencing to determine the transcriptomic consequences of pathogenic TNNT2 variants and adapted CRISPR/Cas9 to engineer a transcriptional reporter assay to assist prediction of TNNT2 variant pathogenicity. We also studied variant-specific pathophysiology using a thin filament-directed calcium reporter to monitor changes in myofilament calcium affinity. RESULTS Hypertrophic cardiomyopathy-associated TNNT2 variants caused increased cardiac microtissue contraction, whereas dilated cardiomyopathy-associated variants decreased contraction. TNNT2 variant-dependent changes in sarcomere contractile function induced graded regulation of 101 gene transcripts, including MAPK (mitogen-activated protein kinase) signaling targets, HOPX, and NPPB. We distinguished pathogenic TNNT2 variants from wildtype controls using a sarcomere functional reporter engineered by inserting tdTomato into the endogenous NPPB locus. On the basis of a combination of NPPB reporter activity and cardiac microtissue contraction, our study provides experimental support for the reclassification of 2 pathogenic/likely pathogenic variants and 2 variants of uncertain significance. CONCLUSIONS Our study found that hypertrophic cardiomyopathy-associated TNNT2 variants increased cardiac microtissue contraction, whereas dilated cardiomyopathy-associated variants decreased contraction, both of which paralleled changes in myofilament calcium affinity. Transcriptomic changes, including NPPB levels, directly correlated with sarcomere function and can be used to predict TNNT2 variant pathogenicity.
Collapse
Affiliation(s)
| | - Feria A Ladha
- University of Connecticut Health Center (A.M.P., F.A.L., R.R., J.T.H.)
| | - David J Mellert
- The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.)
| | - Nicholas Legere
- The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.)
| | - Rachel Cohn
- The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.)
| | - Robert Romano
- University of Connecticut Health Center (A.M.P., F.A.L., R.R., J.T.H.)
| | - Ketan Thakar
- The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.)
| | - Yu-Sheng Chen
- The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.)
| | - J Travis Hinson
- University of Connecticut Health Center (A.M.P., F.A.L., R.R., J.T.H.).,The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.).,Calhoun Cardiology Center, UConn Health (J.T.H.), Farmington
| |
Collapse
|
26
|
Johnson D, Landim-Vieira M, Solı S C, Zhu L, Robinson JM, Pinto JR, Chalovich JM. Eliminating the First Inactive State and Stabilizing the Active State of the Cardiac Regulatory System Alters Behavior in Solution and in Ordered Systems. Biochemistry 2020; 59:3487-3497. [PMID: 32840354 DOI: 10.1021/acs.biochem.0c00430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Calcium binding to troponin C (TnC) is insufficient for full activation of myosin ATPase activity by actin-tropomyosin-troponin. Previous attempts to investigate full activation utilized ATP-free myosin or chemically modified myosin to stabilize the active state of regulated actin. We utilized the Δ14-TnT and the A8V-TnC mutants to stabilize the activated state at saturating Ca2+ and to eliminate one of the inactive states at low Ca2+. The observed effects differed in solution studies and in the more ordered in vitro motility assay and in skinned cardiac muscle preparations. At saturating Ca2+, full activation with Δ14-TnT·A8V-TnC decreased the apparent KM for actin-activated ATPase activity compared to bare actin filaments. Rates of in vitro motility increased at both high and low Ca2+ with Δ14-TnT; the maximum shortening speed at high Ca2+ increased 1.8-fold. Cardiac muscle preparations exhibited increased Ca2+ sensitivity and large increases in resting force with either Δ14-TnT or Δ14-TnT·A8V-TnC. We also observed a significant increase in the maximal rate of tension redevelopment. The results of full activation with Ca2+ and Δ14-TnT·A8V-TnC confirmed and extended several earlier observations using other means of reaching full activation. Furthermore, at low Ca2+, elimination of the first inactive state led to partial activation. This work also confirms, in three distinct experimental systems, that troponin is able to stabilize the active state of actin-tropomyosin-troponin without the need for high-affinity myosin binding. The results are relevant to the reason for two inactive states and for the role of force producing myosin in regulation.
Collapse
Affiliation(s)
- Dylan Johnson
- Department of Biochemistry & Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, North Carolina, United States
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Christopher Solı S
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota 57007, United States
| | - Li Zhu
- Department of Biochemistry & Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, North Carolina, United States
| | - John M Robinson
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota 57007, United States
| | - Jose R Pinto
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Joseph M Chalovich
- Department of Biochemistry & Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, North Carolina, United States
| |
Collapse
|
27
|
Alimadadi A, Manandhar I, Aryal S, Munroe PB, Joe B, Cheng X. Machine learning-based classification and diagnosis of clinical cardiomyopathies. Physiol Genomics 2020; 52:391-400. [PMID: 32744882 DOI: 10.1152/physiolgenomics.00063.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Dilated cardiomyopathy (DCM) and ischemic cardiomyopathy (ICM) are two common types of cardiomyopathies leading to heart failure. Accurate diagnostic classification of different types of cardiomyopathies is critical for precision medicine in clinical practice. In this study, we hypothesized that machine learning (ML) can be used as a novel diagnostic approach to analyze cardiac transcriptomic data for classifying clinical cardiomyopathies. RNA-Seq data of human left ventricle tissues were collected from 41 DCM patients, 47 ICM patients, and 49 nonfailure controls (NF) and tested using five ML algorithms: support vector machine with radial kernel (svmRadial), neural networks with principal component analysis (pcaNNet), decision tree (DT), elastic net (ENet), and random forest (RF). Initial ML classifications achieved ~93% accuracy (svmRadial) for NF vs. DCM, ~82% accuracy (RF) for NF vs. ICM, and ~80% accuracy (ENet and svmRadial) for DCM vs. ICM. Next, 50 highly contributing genes (HCGs) for classifying NF and DCM, 68 HCGs for classifying NF and ICM, and 59 HCGs for classifying DCM and ICM were selected for retraining ML models. Impressively, the retrained models achieved ~90% accuracy (RF) for NF vs. DCM, ~90% accuracy (pcaNNet) for NF vs. ICM, and ~85% accuracy (pcaNNet and RF) for DCM vs. ICM. Pathway analyses further confirmed the involvement of those selected HCGs in cardiac dysfunctions such as cardiomyopathies, cardiac hypertrophies, and fibrosis. Overall, our study demonstrates the promising potential of using artificial intelligence via ML modeling as a novel approach to achieve a greater level of precision in diagnosing different types of cardiomyopathies.
Collapse
Affiliation(s)
- Ahmad Alimadadi
- Bioinformatics & Artificial Intelligence Laboratory, Center for Hypertension and Precision Medicine, Program in Physiological Genomics, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio.,Bioinformatics Program, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Ishan Manandhar
- Bioinformatics & Artificial Intelligence Laboratory, Center for Hypertension and Precision Medicine, Program in Physiological Genomics, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio.,Bioinformatics Program, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Sachin Aryal
- Bioinformatics & Artificial Intelligence Laboratory, Center for Hypertension and Precision Medicine, Program in Physiological Genomics, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio.,Bioinformatics Program, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute & National Institute of Health Research Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Bina Joe
- Bioinformatics & Artificial Intelligence Laboratory, Center for Hypertension and Precision Medicine, Program in Physiological Genomics, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio.,Bioinformatics Program, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Xi Cheng
- Bioinformatics & Artificial Intelligence Laboratory, Center for Hypertension and Precision Medicine, Program in Physiological Genomics, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| |
Collapse
|
28
|
Strianese O, Rizzo F, Ciccarelli M, Galasso G, D’Agostino Y, Salvati A, Del Giudice C, Tesorio P, Rusciano MR. Precision and Personalized Medicine: How Genomic Approach Improves the Management of Cardiovascular and Neurodegenerative Disease. Genes (Basel) 2020; 11:E747. [PMID: 32640513 PMCID: PMC7397223 DOI: 10.3390/genes11070747] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Life expectancy has gradually grown over the last century. This has deeply affected healthcare costs, since the growth of an aging population is correlated to the increasing burden of chronic diseases. This represents the interesting challenge of how to manage patients with chronic diseases in order to improve health care budgets. Effective primary prevention could represent a promising route. To this end, precision, together with personalized medicine, are useful instruments in order to investigate pathological processes before the appearance of clinical symptoms and to guide physicians to choose a targeted therapy to manage the patient. Cardiovascular and neurodegenerative diseases represent suitable models for taking full advantage of precision medicine technologies applied to all stages of disease development. The availability of high technology incorporating artificial intelligence and advancement progress made in the field of biomedical research have been substantial to understand how genes, epigenetic modifications, aging, nutrition, drugs, microbiome and other environmental factors can impact health and chronic disorders. The aim of the present review is to address how precision and personalized medicine can bring greater clarity to the clinical and biological complexity of these types of disorders associated with high mortality, involving tremendous health care costs, by describing in detail the methods that can be applied. This might offer precious tools for preventive strategies and possible clues on the evolution of the disease and could help in predicting morbidity, mortality and detecting chronic disease indicators much earlier in the disease course. This, of course, will have a major effect on both improving the quality of care and quality of life of the patients and reducing time efforts and healthcare costs.
Collapse
Affiliation(s)
- Oriana Strianese
- Clinical Research and Innovation, Clinica Montevergine S.p.A., 83013 Mercogliano, Italy; (O.S.); (C.D.G.)
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (F.R.); (Y.D.); (A.S.)
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (F.R.); (Y.D.); (A.S.)
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (M.C.); (G.G.)
| | - Gennaro Galasso
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (M.C.); (G.G.)
| | - Ylenia D’Agostino
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (F.R.); (Y.D.); (A.S.)
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (F.R.); (Y.D.); (A.S.)
| | - Carmine Del Giudice
- Clinical Research and Innovation, Clinica Montevergine S.p.A., 83013 Mercogliano, Italy; (O.S.); (C.D.G.)
| | - Paola Tesorio
- Unit of Cardiology, Clinica Montevergine S.p.A., 83013 Mercogliano, Italy;
| | - Maria Rosaria Rusciano
- Clinical Research and Innovation, Clinica Montevergine S.p.A., 83013 Mercogliano, Italy; (O.S.); (C.D.G.)
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (M.C.); (G.G.)
| |
Collapse
|
29
|
Cassani M, Fernandes S, Vrbsky J, Ergir E, Cavalieri F, Forte G. Combining Nanomaterials and Developmental Pathways to Design New Treatments for Cardiac Regeneration: The Pulsing Heart of Advanced Therapies. Front Bioeng Biotechnol 2020; 8:323. [PMID: 32391340 PMCID: PMC7193099 DOI: 10.3389/fbioe.2020.00323] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
The research for heart therapies is challenged by the limited intrinsic regenerative capacity of the adult heart. Moreover, it has been hampered by the poor results obtained by tissue engineering and regenerative medicine attempts at generating functional beating constructs able to integrate with the host tissue. For this reason, organ transplantation remains the elective treatment for end-stage heart failure, while novel strategies aiming to promote cardiac regeneration or repair lag behind. The recent discovery that adult cardiomyocytes can be ectopically induced to enter the cell cycle and proliferate by a combination of microRNAs and cardioprotective drugs, like anti-oxidant, anti-inflammatory, anti-coagulants and anti-platelets agents, fueled the quest for new strategies suited to foster cardiac repair. While proposing a revolutionary approach for heart regeneration, these studies raised serious issues regarding the efficient controlled delivery of the therapeutic cargo, as well as its timely removal or metabolic inactivation from the site of action. Especially, there is need for innovative treatment because of evidence of severe side effects caused by pleiotropic drugs. Biocompatible nanoparticles possess unique physico-chemical properties that have been extensively exploited for overcoming the limitations of standard medical therapies. Researchers have put great efforts into the optimization of the nanoparticles synthesis and functionalization, to control their interactions with the biological milieu and use as a viable alternative to traditional approaches. Nanoparticles can be used for diagnosis and deliver therapies in a personalized and targeted fashion. Regarding the treatment of cardiovascular diseases, nanoparticles-based strategies have provided very promising outcomes, in preclinical studies, during the last years. Efficient encapsulation of a large variety of cargos, specific release at the desired site and improvement of cardiac function are some of the main achievements reached so far by nanoparticle-based treatments in animal models. This work offers an overview on the recent nanomedical applications for cardiac regeneration and highlights how the versatility of nanomaterials can be combined with the newest molecular biology discoveries to advance cardiac regeneration therapies.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Soraia Fernandes
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Jan Vrbsky
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Ece Ergir
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria
| | - Francesca Cavalieri
- School of Science, RMIT University, Melbourne, VIC, Australia
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma “Tor Vergata”, Via Della Ricerca Scientifica, Rome, Italy
| | - Giancarlo Forte
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| |
Collapse
|
30
|
Towbin JA. Genetic arrhythmias complicating patients with dilated cardiomyopathy: How it happens. Heart Rhythm 2020; 17:313-314. [PMID: 31634619 DOI: 10.1016/j.hrthm.2019.10.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Jeffrey A Towbin
- Heart Institute, Division of Pediatric Cardiology, Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, Tennessee.
| |
Collapse
|
31
|
Clippinger SR, Cloonan PE, Greenberg L, Ernst M, Stump WT, Greenberg MJ. Disrupted mechanobiology links the molecular and cellular phenotypes in familial dilated cardiomyopathy. Proc Natl Acad Sci U S A 2019; 116:17831-17840. [PMID: 31427533 PMCID: PMC6731759 DOI: 10.1073/pnas.1910962116] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Familial dilated cardiomyopathy (DCM) is a leading cause of sudden cardiac death and a major indicator for heart transplant. The disease is frequently caused by mutations of sarcomeric proteins; however, it is not well understood how these molecular mutations lead to alterations in cellular organization and contractility. To address this critical gap in our knowledge, we studied the molecular and cellular consequences of a DCM mutation in troponin-T, ΔK210. We determined the molecular mechanism of ΔK210 and used computational modeling to predict that the mutation should reduce the force per sarcomere. In mutant cardiomyocytes, we found that ΔK210 not only reduces contractility but also causes cellular hypertrophy and impairs cardiomyocytes' ability to adapt to changes in substrate stiffness (e.g., heart tissue fibrosis that occurs with aging and disease). These results help link the molecular and cellular phenotypes and implicate alterations in mechanosensing as an important factor in the development of DCM.
Collapse
Affiliation(s)
- Sarah R Clippinger
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Paige E Cloonan
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Lina Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Melanie Ernst
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - W Tom Stump
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
32
|
Peña-Peña ML, Monserrat L. Papel de la genética en la estratificación del riesgo de pacientes con miocardiopatía dilatada no isquémica. Rev Esp Cardiol 2019. [DOI: 10.1016/j.recesp.2018.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
33
|
Moving beyond simple answers to complex disorders in sarcomeric cardiomyopathies: the role of integrated systems. Pflugers Arch 2019; 471:661-671. [PMID: 30848350 DOI: 10.1007/s00424-019-02269-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 12/26/2022]
Abstract
The classic clinical definition of hypertrophic cardiomyopathy (HCM) as originally described by Teare is deceptively simple, "left ventricular hypertrophy in the absence of any identifiable cause." Longitudinal studies, however, including a seminal study performed by Frank and Braunwald in 1968, clearly described the disorder much as we know it today, a complex, progressive, and highly variable cardiomyopathy affecting ~ 1/500 individuals worldwide. Subsequent genetic linkage studies in the early 1990s identified mutations in virtually all of the protein components of the cardiac sarcomere as the primary molecular cause of HCM. In addition, a substantial proportion of inherited dilated cardiomyopathy (DCM) has also been linked to sarcomeric protein mutations. Despite our deep understanding of the overall function of the sarcomere as the primary driver of cardiac contractility, the ability to use genotype in patient management remains elusive. A persistent challenge in the field from both the biophysical and clinical standpoints is how to rigorously link high-resolution protein dynamics and mechanics to the long-term cardiovascular remodeling process that characterizes these complex disorders. In this review, we will explore the depth of the problem from both the standpoint of a multi-subunit, highly conserved and dynamic "machine" to the resultant clinical and structural human phenotype with an emphasis on new, integrative approaches that can be widely applied to identify both novel disease mechanisms and new therapeutic targets for these primary biophysical disorders of the cardiac sarcomere.
Collapse
|
34
|
Peña-Peña ML, Monserrat L. Risk Stratification in Patients With Nonisquemic Dilated Cardiomyopathy. The Role of Genetic Testing. ACTA ACUST UNITED AC 2019; 72:333-340. [PMID: 30792015 DOI: 10.1016/j.rec.2018.10.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/24/2018] [Indexed: 12/28/2022]
Abstract
Dilated cardiomyopathy is inherited in nearly 50% of cases. More than 90 genes have been associated with this disease, which is one of the main causes of heart transplant and has been associated with an increased risk of sudden cardiac death. Risk stratification in these patients continues to be challenging. The identification of the specific etiology of the disease is very useful for the early detection of mutation carriers. Genetic study often provides prognostic information and can determine the therapeutic approach. Wide phenotypic variability is observed depending on the mutated gene, the type of mutation, and the presence of additional genetic and environmental factors.
Collapse
Affiliation(s)
- Maria Luisa Peña-Peña
- Unidad de Cardiopatías Familiares, Departamento de Cardiología, Hospital Universitario Virgen del Rocío, Seville, Spain; Departamento de Cardiología, Health in Code, A Coruña, Spain.
| | | |
Collapse
|
35
|
van der Velden J, Stienen GJM. Cardiac Disorders and Pathophysiology of Sarcomeric Proteins. Physiol Rev 2019; 99:381-426. [PMID: 30379622 DOI: 10.1152/physrev.00040.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The sarcomeric proteins represent the structural building blocks of heart muscle, which are essential for contraction and relaxation. During recent years, it has become evident that posttranslational modifications of sarcomeric proteins, in particular phosphorylation, tune cardiac pump function at rest and during exercise. This delicate, orchestrated interaction is also influenced by mutations, predominantly in sarcomeric proteins, which cause hypertrophic or dilated cardiomyopathy. In this review, we follow a bottom-up approach starting from a description of the basic components of cardiac muscle at the molecular level up to the various forms of cardiac disorders at the organ level. An overview is given of sarcomere changes in acquired and inherited forms of cardiac disease and the underlying disease mechanisms with particular reference to human tissue. A distinction will be made between the primary defect and maladaptive/adaptive secondary changes. Techniques used to unravel functional consequences of disease-induced protein changes are described, and an overview of current and future treatments targeted at sarcomeric proteins is given. The current evidence presented suggests that sarcomeres not only form the basis of cardiac muscle function but also represent a therapeutic target to combat cardiac disease.
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Ger J M Stienen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| |
Collapse
|
36
|
Schultheiss HP, Fairweather D, Caforio ALP, Escher F, Hershberger RE, Lipshultz SE, Liu PP, Matsumori A, Mazzanti A, McMurray J, Priori SG. Dilated cardiomyopathy. Nat Rev Dis Primers 2019; 5:32. [PMID: 31073128 PMCID: PMC7096917 DOI: 10.1038/s41572-019-0084-1] [Citation(s) in RCA: 351] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Dilated cardiomyopathy (DCM) is a clinical diagnosis characterized by left ventricular or biventricular dilation and impaired contraction that is not explained by abnormal loading conditions (for example, hypertension and valvular heart disease) or coronary artery disease. Mutations in several genes can cause DCM, including genes encoding structural components of the sarcomere and desmosome. Nongenetic forms of DCM can result from different aetiologies, including inflammation of the myocardium due to an infection (mostly viral); exposure to drugs, toxins or allergens; and systemic endocrine or autoimmune diseases. The heterogeneous aetiology and clinical presentation of DCM make a correct and timely diagnosis challenging. Echocardiography and other imaging techniques are required to assess ventricular dysfunction and adverse myocardial remodelling, and immunological and histological analyses of an endomyocardial biopsy sample are indicated when inflammation or infection is suspected. As DCM eventually leads to impaired contractility, standard approaches to prevent or treat heart failure are the first-line treatment for patients with DCM. Cardiac resynchronization therapy and implantable cardioverter-defibrillators may be required to prevent life-threatening arrhythmias. In addition, identifying the probable cause of DCM helps tailor specific therapies to improve prognosis. An improved aetiology-driven personalized approach to clinical care will benefit patients with DCM, as will new diagnostic tools, such as serum biomarkers, that enable early diagnosis and treatment.
Collapse
Affiliation(s)
- Heinz-Peter Schultheiss
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany. .,Department of Cardiology, Charité-Universitaetsmedizin Berlin, Berlin, Germany.
| | - DeLisa Fairweather
- Mayo Clinic, Department of Cardiovascular Medicine, Jacksonville, FL, USA.
| | - Alida L. P. Caforio
- 0000 0004 1757 3470grid.5608.bDivision of Cardiology, Department of Cardiological Thoracic and Vascular Sciences and Public Health, University of Padua, Padova, Italy
| | - Felicitas Escher
- grid.486773.9Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany ,0000 0001 2218 4662grid.6363.0Department of Cardiology, Charité–Universitaetsmedizin Berlin, Berlin, Germany ,0000 0004 5937 5237grid.452396.fDZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Ray E. Hershberger
- 0000 0001 2285 7943grid.261331.4Divisions of Human Genetics and Cardiovascular Medicine in the Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH USA
| | - Steven E. Lipshultz
- 0000 0004 1936 9887grid.273335.3Department of Pediatrics, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY USA ,0000 0000 9958 7286grid.413993.5Oishei Children’s Hospital, Buffalo, NY USA ,Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Peter P. Liu
- 0000 0001 2182 2255grid.28046.38University of Ottawa Heart Institute, Ottawa, Ontario Canada
| | - Akira Matsumori
- grid.410835.bClinical Research Center, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Andrea Mazzanti
- 0000 0004 1762 5736grid.8982.bDepartment of Molecular Medicine, University of Pavia, Pavia, Italy ,Department of Molecular Cardiology, IRCCS ICS Maugeri, Pavia, Italy
| | - John McMurray
- 0000 0001 2193 314Xgrid.8756.cBritish Heart Foundation (BHF) Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Silvia G. Priori
- 0000 0004 1762 5736grid.8982.bDepartment of Molecular Medicine, University of Pavia, Pavia, Italy ,Department of Molecular Cardiology, IRCCS ICS Maugeri, Pavia, Italy
| |
Collapse
|
37
|
Ly T, Pappas CT, Johnson D, Schlecht W, Colpan M, Galkin VE, Gregorio CC, Dong WJ, Kostyukova AS. Effects of cardiomyopathy-linked mutations K15N and R21H in tropomyosin on thin-filament regulation and pointed-end dynamics. Mol Biol Cell 2018; 30:268-281. [PMID: 30462572 PMCID: PMC6589558 DOI: 10.1091/mbc.e18-06-0406] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Missense mutations K15N and R21H in striated muscle tropomyosin are linked to dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM), respectively. Tropomyosin, together with the troponin complex, regulates muscle contraction and, along with tropomodulin and leiomodin, controls the uniform thin-filament lengths crucial for normal sarcomere structure and function. We used Förster resonance energy transfer to study effects of the tropomyosin mutations on the structure and kinetics of the cardiac troponin core domain associated with the Ca2+-dependent regulation of cardiac thin filaments. We found that the K15N mutation desensitizes thin filaments to Ca2+ and slows the kinetics of structural changes in troponin induced by Ca2+ dissociation from troponin, while the R21H mutation has almost no effect on these parameters. Expression of the K15N mutant in cardiomyocytes decreases leiomodin’s thin-filament pointed-end assembly but does not affect tropomodulin’s assembly at the pointed end. Our in vitro assays show that the R21H mutation causes a twofold decrease in tropomyosin’s affinity for F-actin and affects leiomodin’s function. We suggest that the K15N mutation causes DCM by altering Ca2+-dependent thin-filament regulation and that one of the possible HCM-causing mechanisms by the R21H mutation is through alteration of leiomodin’s function.
Collapse
Affiliation(s)
- Thu Ly
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164
| | - Christopher T Pappas
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721
| | - Dylan Johnson
- Department of Biochemistry and Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, NC 27834
| | - William Schlecht
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164.,Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164
| | - Mert Colpan
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721
| | - Vitold E Galkin
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721
| | - Wen-Ji Dong
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164.,Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164
| | - Alla S Kostyukova
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164
| |
Collapse
|
38
|
Alves ML, Warren CM, Simon JN, Gaffin RD, Montminy EM, Wieczorek DF, Solaro RJ, Wolska BM. Early sensitization of myofilaments to Ca2+ prevents genetically linked dilated cardiomyopathy in mice. Cardiovasc Res 2018; 113:915-925. [PMID: 28379313 DOI: 10.1093/cvr/cvx068] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 03/31/2017] [Indexed: 12/14/2022] Open
Abstract
Background Dilated cardiomoypathies (DCM) are a heterogeneous group of inherited and acquired diseases characterized by decreased contractility and enlargement of cardiac chambers and a major cause of morbidity and mortality. Mice with Glu54Lys mutation in α-tropomyosin (Tm54) demonstrate typical DCM phenotype with reduced myofilament Ca2+ sensitivity. We tested the hypothesis that early sensitization of the myofilaments to Ca2+ in DCM can prevent the DCM phenotype. Methods and results To sensitize Tm54 myofilaments, we used a genetic approach and crossbred Tm54 mice with mice expressing slow skeletal troponin I (ssTnI) that sensitizes myofilaments to Ca2+. Four groups of mice were used: non-transgenic (NTG), Tm54, ssTnI and Tm54/ssTnI (DTG). Systolic function was significantly reduced in the Tm54 mice compared to NTG, but restored in DTG mice. Tm54 mice also showed increased diastolic LV dimensions and HW/BW ratios, when compared to NTG, which were improved in the DTG group. β-myosin heavy chain expression was increased in the Tm54 animals compared to NTG and was partially restored in DTG group. Analysis by 2D-DIGE indicated a significant decrease in two phosphorylated spots of cardiac troponin I (cTnI) in the DTG animals compared to NTG and Tm54. Analysis by 2D-DIGE also indicated no significant changes in troponin T, regulatory light chain, myosin binding protein C and tropomyosin phosphorylation. Conclusion Our data indicate that decreased myofilament Ca2+ sensitivity is an essential element in the pathophysiology of thin filament linked DCM. Sensitization of myofilaments to Ca2+ in the early stage of DCM may be a useful therapeutic strategy in thin filament linked DCM.
Collapse
Affiliation(s)
- Marco L Alves
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA.,Center for Research in Echocardiography and Cardiology, Heart Institute, University of Sao Paulo, Avenida Dr. Eneas de Carvalho Aguiar 44, 05403-900, Sao Paulo, Brazil
| | - Chad M Warren
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA
| | - Jillian N Simon
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA
| | - Robert D Gaffin
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA
| | - Eric M Montminy
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA
| | - David F Wieczorek
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - R John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA
| | - Beata M Wolska
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA.,Department of Medicine, Division of Cardiology, University of Illinois, 840 S Wood St. (M/C 715), Chicago, IL 60612, USA
| |
Collapse
|
39
|
Johnston JR, Chase PB, Pinto JR. Troponin through the looking-glass: emerging roles beyond regulation of striated muscle contraction. Oncotarget 2017; 9:1461-1482. [PMID: 29416706 PMCID: PMC5787451 DOI: 10.18632/oncotarget.22879] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/20/2017] [Indexed: 01/03/2023] Open
Abstract
Troponin is a heterotrimeric Ca2+-binding protein that has a well-established role in regulating striated muscle contraction. However, mounting evidence points to novel cellular functions of troponin, with profound implications in cancer, cardiomyopathy pathogenesis and skeletal muscle aging. Here, we highlight the non-canonical roles and aberrant expression patterns of troponin beyond the sarcomeric milieu. Utilizing bioinformatics tools and online databases, we also provide pathway, subcellular localization, and protein-protein/DNA interaction analyses that support a role for troponin in multiple subcellular compartments. This emerging knowledge challenges the conventional view of troponin as a sarcomere-specific protein exclusively involved in muscle contraction and may transform the way we think about sarcomeric proteins, particularly in the context of human disease and aging.
Collapse
Affiliation(s)
- Jamie R Johnston
- Department of Biomedical Sciences, The Florida State University College of Medicine, Tallahassee, FL, 32306-4300, USA
| | - P Bryant Chase
- Department of Biological Science, The Florida State University, Tallahassee, FL, 32306-4370, USA
| | - Jose Renato Pinto
- Department of Biomedical Sciences, The Florida State University College of Medicine, Tallahassee, FL, 32306-4300, USA
| |
Collapse
|
40
|
Liu HC, Chu TY, Chen LL, Gui WJ, Zhu GN. The cardiovascular toxicity of triadimefon in early life stage of zebrafish and potential implications to human health. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 231:1093-1103. [PMID: 28803741 DOI: 10.1016/j.envpol.2017.05.072] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/24/2017] [Accepted: 05/26/2017] [Indexed: 06/07/2023]
Abstract
The health risk of triadimefon (TF) to cardiovascular system of human is still unclear, especially to pesticide suicides population, occupational population (farmers, retailers and pharmaceutical workers), and special population (young children and infants, pregnant women, older people, and those with compromised immune systems) who are at a greater risk. Therefore, firstly we explored the toxic effects and possible mechanism of cardiovascular toxicity induced by TF using zebrafish model. Zebrafish at stage of 48 h post fertilization (hpf) exposed to TF for 24 h exhibited morphological malformations which were further confirmed by histopathologic examination, including pericardial edema, circulation abnormalities, serious venous thrombosis and increased distance between the sinus venosus (SV) and bulbus arteriosus (BA) regions of the heart. In addition to morphological changes, TF induced functional deficits in the heart of zebrafish, including bradycardia and a significant reduced cardiac output that became more serious at higher concentrations. To better understand the possible molecular mechanisms underlying cardiovascular toxicity in zebrafish, we investigated the transcriptional level of genes related to calcium signaling pathway and cardiac muscle contraction. Q-PCR (quantitative real-time polymerase chain reaction) results demonstrated that the expression level of genes related to ATPase (atp2a1l, atp1b2b, atp1a3b), calcium channel (cacna1ab, cacna1da) and cardiac troponin C (tnnc1a) were significantly decreased after TF exposure. For the first time, the present study revealed that TF exposure had observable morphological and functional negative impacts on cardiovascular system of zebrafish. Mechanistically, this toxicity might result from the pressure of down-regulation of genes associated with calcium signaling pathway and cardiac muscle contraction following TF exposure. These findings generated here can provide information for better pesticide poisoning treatments, occupational disease prevention, and providing theoretical foundation for risk management measures.
Collapse
Affiliation(s)
- Hong-Cui Liu
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| | - Tian-Yi Chu
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| | - Li-Li Chen
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| | - Wen-Jun Gui
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| | - Guo-Nian Zhu
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
41
|
Molecular mechanisms and structural features of cardiomyopathy-causing troponin T mutants in the tropomyosin overlap region. Proc Natl Acad Sci U S A 2017; 114:11115-11120. [PMID: 28973951 DOI: 10.1073/pnas.1710354114] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Point mutations in genes encoding sarcomeric proteins are the leading cause of inherited primary cardiomyopathies. Among them are mutations in the TNNT2 gene that encodes cardiac troponin T (TnT). These mutations are clustered in the tropomyosin (Tm) binding region of TnT, TNT1 (residues 80-180). To understand the mechanistic changes caused by pathogenic mutations in the TNT1 region, six hypertrophic cardiomyopathy (HCM) and two dilated cardiomyopathy (DCM) mutants were studied by biochemical approaches. Binding assays in the absence and presence of actin revealed changes in the affinity of some, but not all, TnT mutants for Tm relative to WT TnT. HCM mutants were hypersensitive and DCM mutants were hyposensitive to Ca2+ in regulated actomyosin ATPase activities. To gain better insight into the disease mechanism, we modeled the structure of TNT1 and its interactions with Tm. The stability predictions made by the model correlated well with the affinity changes observed in vitro of TnT mutants for Tm. The changes in Ca2+ sensitivity showed a strong correlation with the changes in binding affinity. We suggest the primary reason by which these TNNT2 mutations between residues 92 and 144 cause cardiomyopathy is by changing the affinity of TnT for Tm within the TNT1 region.
Collapse
|
42
|
Marian AJ, Braunwald E. Hypertrophic Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy. Circ Res 2017; 121:749-770. [PMID: 28912181 DOI: 10.1161/circresaha.117.311059] [Citation(s) in RCA: 757] [Impact Index Per Article: 108.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetic disorder that is characterized by left ventricular hypertrophy unexplained by secondary causes and a nondilated left ventricle with preserved or increased ejection fraction. It is commonly asymmetrical with the most severe hypertrophy involving the basal interventricular septum. Left ventricular outflow tract obstruction is present at rest in about one third of the patients and can be provoked in another third. The histological features of HCM include myocyte hypertrophy and disarray, as well as interstitial fibrosis. The hypertrophy is also frequently associated with left ventricular diastolic dysfunction. In the majority of patients, HCM has a relatively benign course. However, HCM is also an important cause of sudden cardiac death, particularly in adolescents and young adults. Nonsustained ventricular tachycardia, syncope, a family history of sudden cardiac death, and severe cardiac hypertrophy are major risk factors for sudden cardiac death. This complication can usually be averted by implantation of a cardioverter-defibrillator in appropriate high-risk patients. Atrial fibrillation is also a common complication and is not well tolerated. Mutations in over a dozen genes encoding sarcomere-associated proteins cause HCM. MYH7 and MYBPC3, encoding β-myosin heavy chain and myosin-binding protein C, respectively, are the 2 most common genes involved, together accounting for ≈50% of the HCM families. In ≈40% of HCM patients, the causal genes remain to be identified. Mutations in genes responsible for storage diseases also cause a phenotype resembling HCM (genocopy or phenocopy). The routine applications of genetic testing and preclinical identification of family members represents an important advance. The genetic discoveries have enhanced understanding of the molecular pathogenesis of HCM and have stimulated efforts designed to identify new therapeutic agents.
Collapse
Affiliation(s)
- Ali J Marian
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, Department of Medicine, University of Texas Health Sciences Center at Houston (A.J.M.); Texas Heart Institute, Houston (A.J.M.); and TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (E.B.).
| | - Eugene Braunwald
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, Department of Medicine, University of Texas Health Sciences Center at Houston (A.J.M.); Texas Heart Institute, Houston (A.J.M.); and TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (E.B.)
| |
Collapse
|
43
|
Long PA, Evans JM, Olson TM. Diagnostic Yield of Whole Exome Sequencing in Pediatric Dilated Cardiomyopathy. J Cardiovasc Dev Dis 2017; 4:jcdd4030011. [PMID: 29367541 PMCID: PMC5715713 DOI: 10.3390/jcdd4030011] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/02/2017] [Accepted: 08/04/2017] [Indexed: 12/28/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a heritable, genetically heterogeneous disorder characterized by progressive heart failure. DCM typically remains clinically silent until adulthood, yet symptomatic disease can develop in childhood. We sought to identify the genetic basis of pediatric DCM in 15 sporadic and three affected-siblings cases, comprised of 21 affected children (mean age, five years) whose parents had normal echocardiograms (mean age, 39 years). Twelve underwent cardiac transplantation and five died with severe heart failure. Parent-offspring whole exome sequencing (WES) data were filtered for rare, deleterious, de novo and recessive variants. In prior work, we reported de novo mutations in TNNT2 and RRAGC and compound heterozygous mutations in ALMS1 and TAF1A among four cases in our cohort. Here, de novo mutations in established DCM genes—RBM20, LMNA, TNNT2, and PRDM16—were identified among five additional cases. The RBM20 mutation was previously reported in familial DCM. An identical unreported LMNA mutation was identified in two unrelated cases, both harboring gene-specific defects in cardiomyocyte nuclear morphology. Collectively, WES had a 50% diagnostic yield in our cohort, providing an explanation for pediatric heart failure and enabling informed family planning. Research is ongoing to discover novel DCM genes among the remaining families.
Collapse
Affiliation(s)
- Pamela A Long
- Mayo Graduate School of Biomedical Sciences, Molecular Pharmacology and Experimental Therapeutics Track, Mayo Clinic, Rochester, MN 55905, USA.
| | - Jared M Evans
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA.
| | - Timothy M Olson
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA.
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
44
|
Zhuang Y, Gong YJ, Zhong BF, Zhou Y, Gong L. Bioinformatics method identifies potential biomarkers of dilated cardiomyopathy in a human induced pluripotent stem cell-derived cardiomyocyte model. Exp Ther Med 2017; 14:2771-2778. [PMID: 28912841 DOI: 10.3892/etm.2017.4850] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 02/10/2017] [Indexed: 12/23/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is the most common type of cardiomyopathy that account for the majority of heart failure cases. The present study aimed to reveal the underlying molecular mechanisms of DCM and provide potential biomarkers for detection of this condition. The public dataset of GSE35108 was downloaded, and 4 normal induced pluripotent stem cell (iPSC)-derived cardiomyocytes (N samples) and 4 DCM iPSC-derived cardiomyocytes (DCM samples) were utilized. Raw data were preprocessed, followed by identification of differentially expressed genes (DEGs) between N and DCM samples. Crucial functions and pathway enrichment analysis of DEGs were investigated, and protein-protein interaction (PPI) network analysis was conducted. Furthermore, a module network was extracted from the PPI network, followed by enrichment analysis. A set of 363 DEGs were identified, including 253 upregulated and 110 downregulated genes. Several biological processes (BPs), such as blood vessel development and vasculature development (FLT1 and MMP2), cell adhesion (CDH1, ITGB6, COL6A3, COL6A1 and LAMC2) and extracellular matrix (ECM)-receptor interaction pathway (CDH1, ITGB6, COL6A3, COL6A1 and LAMC2), were significantly enriched by these DEGs. Among them, MMP2, CDH1 and FLT1 were hub nodes in the PPI network, while COL6A3, COL6A1, LAMC2 and ITGB6 were highlighted in module 3 network. In addition, PENK and APLNR were two crucial nodes in module 2, which were linked to each other. In conclusion, several potential biomarkers for DCM were identified, such as MMP2, FLT1, CDH1, ITGB6, COL6A3, COL6A1, LAMC2, PENK and APLNR. These genes may serve significant roles in DCM via involvement of various BPs, such as blood vessel and vasculature development and cell adhesion, and the ECM-receptor interaction pathway.
Collapse
Affiliation(s)
- Yu Zhuang
- Department of Cardiovascular Surgery, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Yu-Jia Gong
- Stomatology Faculty, School of Medicine, Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Bei-Fen Zhong
- Department of Urology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Yi Zhou
- Department of Cardiovascular Surgery, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Li Gong
- Department of Cardiothoracic Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| |
Collapse
|
45
|
England J, Loughna S, Rutland CS. Multiple Species Comparison of Cardiac Troponin T and Dystrophin: Unravelling the DNA behind Dilated Cardiomyopathy. J Cardiovasc Dev Dis 2017; 4:E8. [PMID: 29367539 PMCID: PMC5715711 DOI: 10.3390/jcdd4030008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/01/2017] [Accepted: 07/05/2017] [Indexed: 12/14/2022] Open
Abstract
Animals have frequently been used as models for human disorders and mutations. Following advances in genetic testing and treatment options, and the decreasing cost of these technologies in the clinic, mutations in both companion and commercial animals are now being investigated. A recent review highlighted the genes associated with both human and non-human dilated cardiomyopathy. Cardiac troponin T and dystrophin were observed to be associated with both human and turkey (troponin T) and canine (dystrophin) dilated cardiomyopathies. This review gives an overview of the work carried out in cardiac troponin T and dystrophin to date in both human and animal dilated cardiomyopathy.
Collapse
Affiliation(s)
- Jennifer England
- School of Life Sciences, Medical School, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Siobhan Loughna
- School of Life Sciences, Medical School, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Catrin Sian Rutland
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Sutton Bonington, Leicestershire LE12 5RD, UK.
| |
Collapse
|
46
|
Liu Q, Jiang C, Xu J, Zhao MT, Van Bortle K, Cheng X, Wang G, Chang HY, Wu JC, Snyder MP. Genome-Wide Temporal Profiling of Transcriptome and Open Chromatin of Early Cardiomyocyte Differentiation Derived From hiPSCs and hESCs. Circ Res 2017; 121:376-391. [PMID: 28663367 DOI: 10.1161/circresaha.116.310456] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 06/21/2017] [Accepted: 06/28/2017] [Indexed: 01/13/2023]
Abstract
RATIONALE Recent advances have improved our ability to generate cardiomyocytes from human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs). However, our understanding of the transcriptional regulatory networks underlying early stages (ie, from mesoderm to cardiac mesoderm) of cardiomyocyte differentiation remains limited. OBJECTIVE To characterize transcriptome and chromatin accessibility during early cardiomyocyte differentiation from hiPSCs and hESCs. METHODS AND RESULTS We profiled the temporal changes in transcriptome and chromatin accessibility at genome-wide levels during cardiomyocyte differentiation derived from 2 hiPSC lines and 2 hESC lines at 4 stages: pluripotent stem cells, mesoderm, cardiac mesoderm, and differentiated cardiomyocytes. Overall, RNA sequencing analysis revealed that transcriptomes during early cardiomyocyte differentiation were highly concordant between hiPSCs and hESCs, and clustering of 4 cell lines within each time point demonstrated that changes in genome-wide chromatin accessibility were similar across hiPSC and hESC cell lines. Weighted gene co-expression network analysis (WGCNA) identified several modules that were strongly correlated with different stages of cardiomyocyte differentiation. Several novel genes were identified with high weighted connectivity within modules and exhibited coexpression patterns with other genes, including noncoding RNA LINC01124 and uncharacterized RNA AK127400 in the module related to the mesoderm stage; E-box-binding homeobox 1 (ZEB1) in the module correlated with postcardiac mesoderm. We further demonstrated that ZEB1 is required for early cardiomyocyte differentiation. In addition, based on integrative analysis of both WGCNA and transcription factor motif enrichment analysis, we determined numerous transcription factors likely to play important roles at different stages during cardiomyocyte differentiation, such as T and eomesodermin (EOMES; mesoderm), lymphoid enhancer-binding factor 1 (LEF1) and mesoderm posterior BHLH transcription factor 1 (MESP1; from mesoderm to cardiac mesoderm), meis homeobox 1 (MEIS1) and GATA-binding protein 4 (GATA4) (postcardiac mesoderm), JUN and FOS families, and MEIS2 (cardiomyocyte). CONCLUSIONS Both hiPSCs and hESCs share similar transcriptional regulatory mechanisms underlying early cardiac differentiation, and our results have revealed transcriptional regulatory networks and new factors (eg, ZEB1) controlling early stages of cardiomyocyte differentiation.
Collapse
Affiliation(s)
- Qing Liu
- From the Department of Genetics (Q.L., C.J., K.V.B., M.P.S.), Center for Personal Dynamic Regulomes (J.X., H.Y.C.), Stanford Cardiovascular Institute (M.T.Z., J.C.W.), and Stem Cell Core Facility, Department of Genetics (X.C., G.W.), Stanford University School of Medicine, CA
| | - Chao Jiang
- From the Department of Genetics (Q.L., C.J., K.V.B., M.P.S.), Center for Personal Dynamic Regulomes (J.X., H.Y.C.), Stanford Cardiovascular Institute (M.T.Z., J.C.W.), and Stem Cell Core Facility, Department of Genetics (X.C., G.W.), Stanford University School of Medicine, CA
| | - Jin Xu
- From the Department of Genetics (Q.L., C.J., K.V.B., M.P.S.), Center for Personal Dynamic Regulomes (J.X., H.Y.C.), Stanford Cardiovascular Institute (M.T.Z., J.C.W.), and Stem Cell Core Facility, Department of Genetics (X.C., G.W.), Stanford University School of Medicine, CA
| | - Ming-Tao Zhao
- From the Department of Genetics (Q.L., C.J., K.V.B., M.P.S.), Center for Personal Dynamic Regulomes (J.X., H.Y.C.), Stanford Cardiovascular Institute (M.T.Z., J.C.W.), and Stem Cell Core Facility, Department of Genetics (X.C., G.W.), Stanford University School of Medicine, CA
| | - Kevin Van Bortle
- From the Department of Genetics (Q.L., C.J., K.V.B., M.P.S.), Center for Personal Dynamic Regulomes (J.X., H.Y.C.), Stanford Cardiovascular Institute (M.T.Z., J.C.W.), and Stem Cell Core Facility, Department of Genetics (X.C., G.W.), Stanford University School of Medicine, CA
| | - Xun Cheng
- From the Department of Genetics (Q.L., C.J., K.V.B., M.P.S.), Center for Personal Dynamic Regulomes (J.X., H.Y.C.), Stanford Cardiovascular Institute (M.T.Z., J.C.W.), and Stem Cell Core Facility, Department of Genetics (X.C., G.W.), Stanford University School of Medicine, CA
| | - Guangwen Wang
- From the Department of Genetics (Q.L., C.J., K.V.B., M.P.S.), Center for Personal Dynamic Regulomes (J.X., H.Y.C.), Stanford Cardiovascular Institute (M.T.Z., J.C.W.), and Stem Cell Core Facility, Department of Genetics (X.C., G.W.), Stanford University School of Medicine, CA
| | - Howard Y Chang
- From the Department of Genetics (Q.L., C.J., K.V.B., M.P.S.), Center for Personal Dynamic Regulomes (J.X., H.Y.C.), Stanford Cardiovascular Institute (M.T.Z., J.C.W.), and Stem Cell Core Facility, Department of Genetics (X.C., G.W.), Stanford University School of Medicine, CA
| | - Joseph C Wu
- From the Department of Genetics (Q.L., C.J., K.V.B., M.P.S.), Center for Personal Dynamic Regulomes (J.X., H.Y.C.), Stanford Cardiovascular Institute (M.T.Z., J.C.W.), and Stem Cell Core Facility, Department of Genetics (X.C., G.W.), Stanford University School of Medicine, CA
| | - Michael P Snyder
- From the Department of Genetics (Q.L., C.J., K.V.B., M.P.S.), Center for Personal Dynamic Regulomes (J.X., H.Y.C.), Stanford Cardiovascular Institute (M.T.Z., J.C.W.), and Stem Cell Core Facility, Department of Genetics (X.C., G.W.), Stanford University School of Medicine, CA.
| |
Collapse
|
47
|
Bollen IAE, Schuldt M, Harakalova M, Vink A, Asselbergs FW, Pinto JR, Krüger M, Kuster DWD, van der Velden J. Genotype-specific pathogenic effects in human dilated cardiomyopathy. J Physiol 2017; 595:4677-4693. [PMID: 28436080 PMCID: PMC5509872 DOI: 10.1113/jp274145] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/18/2017] [Indexed: 01/09/2023] Open
Abstract
KEY POINTS Mutations in genes encoding cardiac troponin I (TNNI3) and cardiac troponin T (TNNT2) caused altered troponin protein stoichiometry in patients with dilated cardiomyopathy. TNNI3p.98trunc resulted in haploinsufficiency, increased Ca2+ -sensitivity and reduced length-dependent activation. TNNT2p.K217del caused increased passive tension. A mutation in the gene encoding Lamin A/C (LMNAp.R331Q ) led to reduced maximal force development through secondary disease remodelling in patients suffering from dilated cardiomyopathy. Our study shows that different gene mutations induce dilated cardiomyopathy via diverse cellular pathways. ABSTRACT Dilated cardiomyopathy (DCM) can be caused by mutations in sarcomeric and non-sarcomeric genes. In this study we defined the pathogenic effects of three DCM-causing mutations: the sarcomeric mutations in genes encoding cardiac troponin I (TNNI3p.98truncation ) and cardiac troponin T (TNNT2p.K217deletion ; also known as the p.K210del) and the non-sarcomeric gene mutation encoding lamin A/C (LMNAp.R331Q ). We assessed sarcomeric protein expression and phosphorylation and contractile behaviour in single membrane-permeabilized cardiomyocytes in human left ventricular heart tissue. Exchange with recombinant troponin complex was used to establish the direct pathogenic effects of the mutations in TNNI3 and TNNT2. The TNNI3p.98trunc and TNNT2p.K217del mutation showed reduced expression of troponin I to 39% and 51%, troponin T to 64% and 53%, and troponin C to 73% and 97% of controls, respectively, and altered stoichiometry between the three cardiac troponin subunits. The TNNI3p.98trunc showed pure haploinsufficiency, increased Ca2+ -sensitivity and impaired length-dependent activation. The TNNT2p.K217del mutation showed a significant increase in passive tension that was not due to changes in titin isoform composition or phosphorylation. Exchange with wild-type troponin complex corrected troponin protein levels to 83% of controls in the TNNI3p.98trunc sample. Moreover, upon exchange all functional deficits in the TNNI3p.98trunc and TNNT2p.K217del samples were normalized to control values confirming the pathogenic effects of the troponin mutations. The LMNAp.R331Q mutation resulted in reduced maximal force development due to disease remodelling. Our study shows that different gene mutations induce DCM via diverse cellular pathways.
Collapse
Affiliation(s)
- Ilse A E Bollen
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, the Netherlands
| | - Maike Schuldt
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, the Netherlands
| | - Magdalena Harakalova
- Department of Cardiology, Division of Heart and Lungs, University of Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Division of Heart and Lungs, University of Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands.,Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, the Netherlands.,Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK
| | - Jose R Pinto
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Martina Krüger
- Institute of Cardiovascular Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Diederik W D Kuster
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, the Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, the Netherlands.,Netherlands Heart Institute, Utrecht, the Netherlands
| |
Collapse
|
48
|
Dewan S, McCabe KJ, Regnier M, McCulloch AD. Insights and Challenges of Multi-Scale Modeling of Sarcomere Mechanics in cTn and Tm DCM Mutants-Genotype to Cellular Phenotype. Front Physiol 2017; 8:151. [PMID: 28352236 PMCID: PMC5348544 DOI: 10.3389/fphys.2017.00151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/24/2017] [Indexed: 01/18/2023] Open
Abstract
Dilated Cardiomyopathy (DCM) is a leading cause of sudden cardiac death characterized by impaired pump function and dilatation of cardiac ventricles. In this review we discuss various in silico approaches to elucidating the mechanisms of genetic mutations leading to DCM. The approaches covered in this review focus on bridging the spatial and temporal gaps that exist between molecular and cellular processes. Mutations in sarcomeric regulatory thin filament proteins such as the troponin complex (cTn) and Tropomyosin (Tm) have been associated with DCM. Despite the experimentally-observed myofilament measures of contractility in the case of these mutations, the mechanisms by which the underlying molecular changes and protein interactions scale up to organ failure by these mutations remains elusive. The review highlights multi-scale modeling approaches and their applicability to study the effects of sarcomeric gene mutations in-silico. We discuss some of the insights that can be gained from computational models of cardiac biomechanics when scaling from molecular states to cellular level.
Collapse
Affiliation(s)
- Sukriti Dewan
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kimberly J McCabe
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Michael Regnier
- Departments of Bioengineering and Medicine, University of Washington Seattle, WA, USA
| | - Andrew D McCulloch
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
49
|
Karakikes I, Termglinchan V, Cepeda DA, Lee J, Diecke S, Hendel A, Itzhaki I, Ameen M, Shrestha R, Wu H, Ma N, Shao NY, Seeger T, Woo N, Wilson KD, Matsa E, Porteus MH, Sebastiano V, Wu JC. A Comprehensive TALEN-Based Knockout Library for Generating Human-Induced Pluripotent Stem Cell-Based Models for Cardiovascular Diseases. Circ Res 2017; 120:1561-1571. [PMID: 28246128 DOI: 10.1161/circresaha.116.309948] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 02/23/2017] [Accepted: 02/28/2017] [Indexed: 12/21/2022]
Abstract
RATIONALE Targeted genetic engineering using programmable nucleases such as transcription activator-like effector nucleases (TALENs) is a valuable tool for precise, site-specific genetic modification in the human genome. OBJECTIVE The emergence of novel technologies such as human induced pluripotent stem cells (iPSCs) and nuclease-mediated genome editing represent a unique opportunity for studying cardiovascular diseases in vitro. METHODS AND RESULTS By incorporating extensive literature and database searches, we designed a collection of TALEN constructs to knockout 88 human genes that are associated with cardiomyopathies and congenital heart diseases. The TALEN pairs were designed to induce double-strand DNA break near the starting codon of each gene that either disrupted the start codon or introduced a frameshift mutation in the early coding region, ensuring faithful gene knockout. We observed that all the constructs were active and disrupted the target locus at high frequencies. To illustrate the utility of the TALEN-mediated knockout technique, 6 individual genes (TNNT2, LMNA/C, TBX5, MYH7, ANKRD1, and NKX2.5) were knocked out with high efficiency and specificity in human iPSCs. By selectively targeting a pathogenic mutation (TNNT2 p.R173W) in patient-specific iPSC-derived cardiac myocytes, we demonstrated that the knockout strategy ameliorates the dilated cardiomyopathy phenotype in vitro. In addition, we modeled the Holt-Oram syndrome in iPSC-cardiac myocytes in vitro and uncovered novel pathways regulated by TBX5 in human cardiac myocyte development. CONCLUSIONS Collectively, our study illustrates the powerful combination of iPSCs and genome editing technologies for understanding the biological function of genes, and the pathological significance of genetic variants in human cardiovascular diseases. The methods, strategies, constructs, and iPSC lines developed in this study provide a validated, readily available resource for cardiovascular research.
Collapse
Affiliation(s)
- Ioannis Karakikes
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Vittavat Termglinchan
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Diana A Cepeda
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Jaecheol Lee
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Sebastian Diecke
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Ayal Hendel
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Ilanit Itzhaki
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Mohamed Ameen
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Rajani Shrestha
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Haodi Wu
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Ning Ma
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Ning-Yi Shao
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Timon Seeger
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Nicole Woo
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Kitchener D Wilson
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Elena Matsa
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Matthew H Porteus
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.)
| | - Vittorio Sebastiano
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.).
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute (I.K., V.T., J.L., S.D., I.I., M.A., R.S., H.W., N.M., N.-Y.S., T.S., N.W., K.D.W., E.M., J.C.W.), Department of Cardiothoracic Surgery (I.K.), Division of Cardiovascular Medicine, Department of Medicine (V.T., J.C.W.), CA; Institute of Stem Cell Biology and Regenerative Medicine (D.A.C., V.S., J.C.W.), Departments of Pediatrics (A.H., M.H.P.), Pathology (K.D.W.), and Obstetrics and Gynecology (V.S.), Stanford University School of Medicine, CA; Berlin Institute of Health, Germany (S.D.); and Max Delbrueck Center, Berlin, Germany (S.D.).
| |
Collapse
|
50
|
Gollapudi SK, Chandra M. Dilated Cardiomyopathy Mutation (R134W) in Mouse Cardiac Troponin T Induces Greater Contractile Deficits against α-Myosin Heavy Chain than against β-Myosin Heavy Chain. Front Physiol 2016; 7:443. [PMID: 27757084 PMCID: PMC5047882 DOI: 10.3389/fphys.2016.00443] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/20/2016] [Indexed: 11/13/2022] Open
Abstract
Many studies have demonstrated that depressed myofilament Ca2+ sensitivity is common to dilated cardiomyopathy (DCM) in humans. However, it remains unclear whether a single determinant-such as myofilament Ca2+ sensitivity-is sufficient to characterize all cases of DCM because the severity of disease varies widely with a given mutation. Because dynamic features dominate in the heart muscle, alterations in dynamic contractile parameters may offer better insight on the molecular mechanisms that underlie disparate effects of DCM mutations on cardiac phenotypes. Dynamic features are dominated by myofilament cooperativity that stem from different sources. One such source is the strong tropomyosin binding region in troponin T (TnT), which is known to modulate crossbridge (XB) recruitment dynamics in a myosin heavy chain (MHC)-dependent manner. Therefore, we hypothesized that the effects of DCM-linked mutations in TnT on contractile dynamics would be differently modulated by α- and β-MHC. After reconstitution with the mouse TnT equivalent (TnTR134W) of the human DCM mutation (R131W), we measured dynamic contractile parameters in detergent-skinned cardiac muscle fiber bundles from normal (α-MHC) and transgenic mice (β-MHC). TnTR134W significantly attenuated the rate constants of tension redevelopment, XB recruitment dynamics, XB distortion dynamics, and the magnitude of length-mediated XB recruitment only in α-MHC fiber bundles. TnTR134W decreased myofilament Ca2+ sensitivity to a greater extent in α-MHC (0.14 pCa units) than in β-MHC fiber bundles (0.08 pCa units). Thus, our data demonstrate that TnTR134W induces a more severe DCM-like contractile phenotype against α-MHC than against β-MHC background.
Collapse
Affiliation(s)
- Sampath K Gollapudi
- Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA
| | - Murali Chandra
- Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA
| |
Collapse
|