1
|
Creso JG, Gokhan I, Rynkiewicz MJ, Lehman W, Moore JR, Campbell SG. In silico and in vitro models reveal the molecular mechanisms of hypocontractility caused by TPM1 M8R. Front Physiol 2024; 15:1452509. [PMID: 39282088 PMCID: PMC11392859 DOI: 10.3389/fphys.2024.1452509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is an inherited disorder often leading to severe heart failure. Linkage studies in affected families have revealed hundreds of different mutations that can cause DCM, with most occurring in genes associated with the cardiac sarcomere. We have developed an investigational pipeline for discovering mechanistic genotype-phenotype relationships in DCM and here apply it to the DCM-linked tropomyosin mutation TPM1 M8R. Atomistic simulations predict that M8R increases flexibility of the tropomyosin chain and enhances affinity for the blocked or inactive state of tropomyosin on actin. Applying these molecular effects to a Markov model of the cardiac thin filament reproduced the shifts in Ca2+sensitivity, maximum force, and a qualitative drop in cooperativity that were observed in an in vitro system containing TPM1 M8R. The model was then used to simulate the impact of M8R expression on twitch contractions of intact cardiac muscle, predicting that M8R would reduce peak force and duration of contraction in a dose-dependent manner. To evaluate this prediction, TPM1 M8R was expressed via adenovirus in human engineered heart tissues and isometric twitch force was observed. The mutant tissues manifested depressed contractility and twitch duration that agreed in detail with model predictions. Additional exploratory simulations suggest that M8R-mediated alterations in tropomyosin-actin interactions contribute more potently than tropomyosin chain stiffness to cardiac twitch dysfunction, and presumably to the ultimate manifestation of DCM. This study is an example of the growing potential for successful in silico prediction of mutation pathogenicity for inherited cardiac muscle disorders.
Collapse
Affiliation(s)
- Jenette G. Creso
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Ilhan Gokhan
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Michael J. Rynkiewicz
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - William Lehman
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Jeffrey R. Moore
- Department of Biological Sciences, University of Massachusetts–Lowell, Lowell, MA, United States
| | - Stuart G. Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
2
|
García-Hernández S, de la Higuera Romero L, Ochoa JP, McKenna WJ. Emerging Themes in Genetics of Hypertrophic Cardiomyopathy: Current Status and Clinical Application. Can J Cardiol 2024; 40:742-753. [PMID: 38244984 DOI: 10.1016/j.cjca.2024.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM), defined clinically by the presence of unexplained left ventricular hypertrophy (LVH), with wall thickness ≥ 1.5 cm, is a phenotype in search of a diagnosis, which is most often a genetically determined, cardiac exclusive, or systemic disorder. Familial evaluation and genetic testing are required for definitive diagnosis. The role of genetic findings in predicting development of disease, outcomes, and increasingly to guide management is evolving with access to larger data sets. The specific mutation and sex of the patient are important determinants that ultimately are likely to guide management. The genetic/familial evaluation is influenced by the accuracy of the clinical diagnosis and the extent/expertise of the genetic laboratory. Genetic testing in a patient with unexplained LVH without systemic manifestations will yield a definite/likely pathogenetic mutation in a sarcomere (30%-50%), regulatory/functional (10%-15%) or metabolic/syndromic (< 5%) gene associated with Mendelian inheritance. The importance of oligo- and polygenic determinants, usually in the absence of Mendelian inheritance, is under investigation with important implications, particularly related to familial evaluation and definition of risk of disease development in relatives of probands. The results of genetic testing are increasingly important in management strategies related to the use of the implantable cardioverter defibrillator for prevention of sudden death, use of myosin inhibitors for refractory symptoms in patients with and without outflow tract obstruction, and-on the immediate horizon-gene therapy. This review will focus on genetic and outcome data in sarcomeric HCM, and minor causative genes with robust evidence of their association will also be considered.
Collapse
Affiliation(s)
| | | | - Juan Pablo Ochoa
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña, A Coruña, Spain; Centro Nacional de Investigaciones Cardiovasculades (CNIC), Madrid, Spain; Health in Code S.L., A Coruña, Spain
| | - William J McKenna
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña, A Coruña, Spain; Institute of Cardiovascular Science, University College London, London, United Kingdom; Health in Code S.L., A Coruña, Spain.
| |
Collapse
|
3
|
Hanel Y, Dittmann S, Müller K, Ioannou ME, Schulze-Bahr E. Case report: cosegregation of a TPM1 in-frame deletion (p.Lys7del) with familial non-compaction cardiomyopathy (NCCM). Clin Res Cardiol 2024; 113:656-660. [PMID: 37067544 PMCID: PMC11026255 DOI: 10.1007/s00392-023-02190-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/24/2023] [Indexed: 04/18/2023]
Affiliation(s)
- Yvonne Hanel
- Department of Cardiovascular Medicine, ERN Reference Center GUARD-Heart, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Albert-Schweitzer-Campus 1 (D3), 48149, Münster, Germany.
| | - Sven Dittmann
- Department of Cardiovascular Medicine, ERN Reference Center GUARD-Heart, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Albert-Schweitzer-Campus 1 (D3), 48149, Münster, Germany
| | - Klara Müller
- Department of Cardiovascular Medicine, ERN Reference Center GUARD-Heart, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Albert-Schweitzer-Campus 1 (D3), 48149, Münster, Germany
| | - Monica Elena Ioannou
- Department of Cardiovascular Medicine, ERN Reference Center GUARD-Heart, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Albert-Schweitzer-Campus 1 (D3), 48149, Münster, Germany
| | - Eric Schulze-Bahr
- Department of Cardiovascular Medicine, ERN Reference Center GUARD-Heart, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Albert-Schweitzer-Campus 1 (D3), 48149, Münster, Germany
| |
Collapse
|
4
|
Pasławska M, Grodzka A, Peczyńska J, Sawicka B, Bossowski AT. Role of miRNA in Cardiovascular Diseases in Children-Systematic Review. Int J Mol Sci 2024; 25:956. [PMID: 38256030 PMCID: PMC10816020 DOI: 10.3390/ijms25020956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
The number of children suffering from cardiovascular diseases (CVDs) is rising globally. Therefore, there is an urgent need to acquire a better understanding of the genetic factors and molecular mechanisms related to the pathogenesis of CVDs in order to develop new prevention and treatment strategies for the future. MicroRNAs (miRNAs) constitute a class of small non-coding RNA fragments that range from 17 to 25 nucleotides in length and play an essential role in regulating gene expression, controlling an abundance of biological aspects of cell life, such as proliferation, differentiation, and apoptosis, thus affecting immune response, stem cell growth, ageing and haematopoiesis. In recent years, the concept of miRNAs as diagnostic markers allowing discrimination between healthy individuals and those affected by CVDs entered the purview of academic debate. In this review, we aimed to systematise available information regarding miRNAs associated with arrhythmias, cardiomyopathies, myocarditis and congenital heart diseases in children. We focused on the targeted genes and metabolic pathways influenced by those particular miRNAs, and finally, tried to determine the future of miRNAs as novel biomarkers of CVD.
Collapse
Affiliation(s)
| | | | | | | | - Artur Tadeusz Bossowski
- Department of Pediatrics, Endocrinology, Diabetology with Cardiology Divisions, Medical University of Bialystok, J. Waszyngtona 17, 15-274 Bialystok, Poland; (M.P.); (A.G.); (J.P.); (B.S.)
| |
Collapse
|
5
|
Kumar P, Paramasivam G, Devasia T, Prabhu M, Rai MK, Prakashini K, Mallya S, Reghunathan D, Megha A, Nayak K, Moka R. A Novel TPM1 Mutation Causes Familial Hypertrophic Cardiomyopathy in an Indian Family: Genetic and Clinical Correlation. Indian J Clin Biochem 2024; 39:142-145. [PMID: 38223010 PMCID: PMC10784234 DOI: 10.1007/s12291-022-01036-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/21/2022] [Indexed: 11/26/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is a common inherited cardiac disorder characterised by unexplained left ventricular hypertrophy in the absence of abnormal loading conditions. The global prevalence of HCM is estimated to be 1 in 250 in the general population. It is caused due to mutations in genes coding for sarcomeric proteins. α-tropomyosin (TPM1) is an important protein in the sarcomeric thin filament which regulates sarcomere contraction. Mutations in TPM1 are known to cause hypertrophic cardiomyopathy, dilated cardiomyopathy and left ventricular non-compaction. Mutations in TPM1 causing hypertrophic cardiomyopathy are < 1%. However, some high-risk mutations causing sudden cardiac death are also known in this gene. We present a case of a novel heterozygous TPM1 mutation, NM_001018005.2:c.203A>G, p.Gln68Arg; co-segregating in an Indian family with hypertrophic cardiomyopathy. Our report expands the mutational spectrum of HCM due to TPM1 and provides the correlated cardiac phenotype.
Collapse
Affiliation(s)
- Prabodh Kumar
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Planetarium Complex, Madhav Nagar, Manipal, 576104 Karnataka India
| | - Ganesh Paramasivam
- Department of Cardiology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Tom Devasia
- Department of Cardiology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Mukund Prabhu
- Department of Cardiology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Maneesh K. Rai
- Department of Cardiology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Mangalore, 575001 Karnataka India
| | - K. Prakashini
- Department of Radiodiagnosis and Imaging, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Sandeep Mallya
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Dinesh Reghunathan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Planetarium Complex, Madhav Nagar, Manipal, 576104 Karnataka India
| | - A. Megha
- Department of Cardiovascular Technology, Manipal College of Health Profession, Manipal Academy of Higher Education (Manipal), Manipal, 576104 Karnataka India
| | - Krishnananda Nayak
- Department of Cardiovascular Technology, Manipal College of Health Profession, Manipal Academy of Higher Education (Manipal), Manipal, 576104 Karnataka India
| | - Rajasekhar Moka
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Planetarium Complex, Madhav Nagar, Manipal, 576104 Karnataka India
| |
Collapse
|
6
|
Matyushenko AM, Nefedova VV, Kochurova AM, Kopylova GV, Koubassova NA, Shestak AG, Yampolskaya DS, Shchepkin DV, Kleymenov SY, Ryabkova NS, Katrukha IA, Bershitsky SY, Zaklyazminskaya EV, Tsaturyan AK, Levitsky DI. Novel Mutation Glu98Lys in Cardiac Tropomyosin Alters Its Structure and Impairs Myocardial Relaxation. Int J Mol Sci 2023; 24:12359. [PMID: 37569730 PMCID: PMC10419091 DOI: 10.3390/ijms241512359] [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: 07/01/2023] [Revised: 07/25/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
We characterized a novel genetic variant c.292G > A (p.E98K) in the TPM1 gene encoding cardiac tropomyosin 1.1 isoform (Tpm1.1), found in a proband with a phenotype of complex cardiomyopathy with conduction dysfunction and slow progressive neuromuscular involvement. To understand the molecular mechanism by which this mutation impairs cardiac function, we produced recombinant Tpm1.1 carrying an E98K substitution and studied how this substitution affects the structure of the Tpm1.1 molecule and its functional properties. The results showed that the E98K substitution in the N-terminal part of the Tpm molecule significantly destabilizes the C-terminal part of Tpm, thus indicating a long-distance destabilizing effect of the substitution on the Tpm coiled-coil structure. The E98K substitution did not noticeably affect Tpm's affinity for F-actin but significantly impaired Tpm's regulatory properties. It increased the Ca2+ sensitivity of the sliding velocity of regulated thin filaments over cardiac myosin in an in vitro motility assay and caused an incomplete block of the thin filament sliding at low Ca2+ concentrations. The incomplete motility block in the absence of Ca2+ can be explained by the loosening of the Tpm interaction with troponin I (TnI), thus increasing Tpm mobility on the surface of an actin filament that partially unlocks the myosin binding sites. This hypothesis is supported by the molecular dynamics (MD) simulation that showed that the E98 Tpm residue is involved in hydrogen bonding with the C-terminal part of TnI. Thus, the results allowed us to explain the mechanism by which the E98K Tpm mutation impairs sarcomeric function and myocardial relaxation.
Collapse
Affiliation(s)
- Alexander M. Matyushenko
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow 119071, Russia; (A.M.M.); (V.V.N.); (D.S.Y.); (S.Y.K.)
| | - Victoria V. Nefedova
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow 119071, Russia; (A.M.M.); (V.V.N.); (D.S.Y.); (S.Y.K.)
| | - Anastasia M. Kochurova
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg 620049, Russia; (A.M.K.); (G.V.K.); (D.V.S.); (S.Y.B.)
| | - Galina V. Kopylova
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg 620049, Russia; (A.M.K.); (G.V.K.); (D.V.S.); (S.Y.B.)
| | - Natalia A. Koubassova
- Institute of Mechanics, Moscow State University, Moscow 119192, Russia; (N.A.K.); (A.K.T.)
| | - Anna G. Shestak
- Petrovsky National Research Centre of Surgery, Moscow 119991, Russia; (A.G.S.); (E.V.Z.)
| | - Daria S. Yampolskaya
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow 119071, Russia; (A.M.M.); (V.V.N.); (D.S.Y.); (S.Y.K.)
| | - Daniil V. Shchepkin
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg 620049, Russia; (A.M.K.); (G.V.K.); (D.V.S.); (S.Y.B.)
| | - Sergey Y. Kleymenov
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow 119071, Russia; (A.M.M.); (V.V.N.); (D.S.Y.); (S.Y.K.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia
| | - Natalia S. Ryabkova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (N.S.R.); (I.A.K.)
- HyTest Ltd., 20520 Turku, Finland
| | - Ivan A. Katrukha
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (N.S.R.); (I.A.K.)
- HyTest Ltd., 20520 Turku, Finland
| | - Sergey Y. Bershitsky
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg 620049, Russia; (A.M.K.); (G.V.K.); (D.V.S.); (S.Y.B.)
| | - Elena V. Zaklyazminskaya
- Petrovsky National Research Centre of Surgery, Moscow 119991, Russia; (A.G.S.); (E.V.Z.)
- N.P. Bochkov Research Centre for Medical Genetics, Moscow 20520, Russia
| | - Andrey K. Tsaturyan
- Institute of Mechanics, Moscow State University, Moscow 119192, Russia; (N.A.K.); (A.K.T.)
| | - Dmitrii I. Levitsky
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow 119071, Russia; (A.M.M.); (V.V.N.); (D.S.Y.); (S.Y.K.)
| |
Collapse
|
7
|
De Novo Asp219Val Mutation in Cardiac Tropomyosin Associated with Hypertrophic Cardiomyopathy. Int J Mol Sci 2022; 24:ijms24010018. [PMID: 36613463 PMCID: PMC9820293 DOI: 10.3390/ijms24010018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM), caused by mutations in thin filament proteins, manifests as moderate cardiac hypertrophy and is associated with sudden cardiac death (SCD). We identified a new de novo variant, c.656A>T (p.D219V), in the TPM1 gene encoding cardiac tropomyosin 1.1 (Tpm) in a young SCD victim with post-mortem-diagnosed HCM. We produced recombinant D219V Tpm1.1 and studied its structural and functional properties using various biochemical and biophysical methods. The D219V mutation did not affect the Tpm affinity for F-actin but increased the thermal stability of the Tpm molecule and Tpm-F-actin complex. The D219V mutation significantly increased the Ca2+ sensitivity of the sliding velocity of thin filaments over cardiac myosin in an in vitro motility assay and impaired the inhibition of the filament sliding at low Ca2+ concentration. The molecular dynamics (MD) simulation provided insight into a possible molecular mechanism of the effect of the mutation that is most likely a cause of the weakening of the Tpm interaction with actin in the "closed" state and so makes it an easier transition to the “open” state. The changes in the Ca2+ regulation of the actin-myosin interaction characteristic of genetic HCM suggest that the mutation is likely pathogenic.
Collapse
|
8
|
Nefedova VV, Kopylova GV, Shchepkin DV, Kochurova AM, Kechko OI, Borzova VA, Ryabkova NS, Katrukha IA, Mitkevich VA, Bershitsky SY, Levitsky DI, Matyushenko AM. Impact of Troponin in Cardiomyopathy Development Caused by Mutations in Tropomyosin. Int J Mol Sci 2022; 23:ijms232415723. [PMID: 36555368 PMCID: PMC9779223 DOI: 10.3390/ijms232415723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Tropomyosin (Tpm) mutations cause inherited cardiac diseases such as hypertrophic and dilated cardiomyopathies. We applied various approaches to investigate the role of cardiac troponin (Tn) and especially the troponin T (TnT) in the pathogenic effects of Tpm cardiomyopathy-associated mutations M8R, K15N, A277V, M281T, and I284V located in the overlap junction of neighboring Tpm dimers. Using co-sedimentation assay and viscosity measurements, we showed that TnT1 (fragment of TnT) stabilizes the overlap junction of Tpm WT and all Tpm mutants studied except Tpm M8R. However, isothermal titration calorimetry (ITC) indicated that TnT1 binds Tpm WT and all Tpm mutants similarly. By using ITC, we measured the direct KD of the Tpm overlap region, N-end, and C-end binding to TnT1. The ITC data revealed that the Tpm C-end binds to TnT1 independently from the N-end, while N-end does not bind. Therefore, we suppose that Tpm M8R binds to TnT1 without forming the overlap junction. We also demonstrated the possible role of Tn isoform composition in the cardiomyopathy development caused by M8R mutation. TnT1 dose-dependently reduced the velocity of F-actin-Tpm filaments containing Tpm WT, Tpm A277V, and Tpm M281T mutants in an in vitro motility assay. All mutations impaired the calcium regulation of the actin-myosin interaction. The M281T and I284V mutations increased the calcium sensitivity, while the K15N and A277V mutations reduced it. The Tpm M8R, M281T, and I284V mutations under-inhibited the velocity at low calcium concentrations. Our results demonstrate that Tpm mutations likely implement their pathogenic effects through Tpm interaction with Tn, cardiac myosin, or other protein partners.
Collapse
Affiliation(s)
- Victoria V. Nefedova
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
- Correspondence:
| | - Galina V. Kopylova
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia
| | - Daniil V. Shchepkin
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia
| | - Anastasia M. Kochurova
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia
| | - Olga I. Kechko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - Vera A. Borzova
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Natalia S. Ryabkova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- HyTest Ltd., 20520 Turku, Finland
| | - Ivan A. Katrukha
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- HyTest Ltd., 20520 Turku, Finland
| | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - Sergey Y. Bershitsky
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia
| | - Dmitrii I. Levitsky
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | | |
Collapse
|
9
|
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: 4.3] [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
|
10
|
Kang JY, Mun D, Chun Y, Kim H, Yun N, Lee SH, Joung B. Generation of a heterozygous TPM1-E192K knock-in human induced pluripotent stem cell line using CRISPR/Cas9 system. Stem Cell Res 2022; 63:102878. [PMID: 35917600 DOI: 10.1016/j.scr.2022.102878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 11/29/2022] Open
Abstract
E192K missense mutation of TPM1 has been found in different types of cardiomyopathies (e.g., hypertrophic cardiomyopathy, dilated cardiomyopathy, and left ventricular non-compaction), leading to systolic dysfunction, diastolic dysfunction, and/or tachyarrhythmias. Here, we generated a heterozygous TPM1-E192K knock-in human induced pluripotent stem cell (iPSC) line using CRISPR/Cas9-based genome editing system. The cells exhibit normal karyotype, typical stem cell morphology, expression of pluripotency markers and differentiation ability into three germ layers. Accordingly, this cell line could provide a useful cell resource for exploring the pathogenic role of TPM1-E192K mutation in different types of cardiomyopathies.
Collapse
Affiliation(s)
- Ji-Young Kang
- Division of Cardiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Dasom Mun
- Division of Cardiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Yumin Chun
- Division of Cardiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Hyoeun Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Nuri Yun
- Institute of Life Science & Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Seung-Hyun Lee
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea.
| | - Boyoung Joung
- Division of Cardiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea.
| |
Collapse
|
11
|
Reindl T, Giese S, Greve JN, Reinke PY, Chizhov I, Latham SL, Mulvihill DP, Taft MH, Manstein DJ. Distinct actin–tropomyosin cofilament populations drive the functional diversification of cytoskeletal myosin motor complexes. iScience 2022; 25:104484. [PMID: 35720262 PMCID: PMC9204724 DOI: 10.1016/j.isci.2022.104484] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/02/2022] [Accepted: 05/24/2022] [Indexed: 11/02/2022] Open
Abstract
The effects of N-terminal acetylation of the high molecular weight tropomyosin isoforms Tpm1.6 and Tpm2.1 and the low molecular weight isoforms Tpm1.12, Tpm3.1, and Tpm4.2 on the actin affinity and the thermal stability of actin-tropomyosin cofilaments are described. Furthermore, we show how the exchange of cytoskeletal tropomyosin isoforms and their N-terminal acetylation affects the kinetic and chemomechanical properties of cytoskeletal actin-tropomyosin-myosin complexes. Our results reveal the extent to which the different actin-tropomyosin-myosin complexes differ in their kinetic and functional properties. The maximum sliding velocity of the actin filament as well as the optimal motor density for continuous unidirectional movement, parameters that were previously considered to be unique and invariant properties of each myosin isoform, are shown to be influenced by the exchange of the tropomyosin isoform and the N-terminal acetylation of tropomyosin. Tpm diversity is largely determined by sequences contributing to the overlap region Global sequence differences are of greater importance than variable exon 6 usage Tpm isoforms confer distinctly altered properties to cytoskeletal myosin motors Cytoskeletal myosins are differentially affected by N-terminal acetylation of Tpm
Collapse
|
12
|
Genetic Insights into Primary Restrictive Cardiomyopathy. J Clin Med 2022; 11:jcm11082094. [PMID: 35456187 PMCID: PMC9027761 DOI: 10.3390/jcm11082094] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/04/2022] Open
Abstract
Restrictive cardiomyopathy is a rare cardiac disease causing severe diastolic dysfunction, ventricular stiffness and dilated atria. In consequence, it induces heart failure often with preserved ejection fraction and is associated with a high mortality. Since it is a poor clinical prognosis, patients with restrictive cardiomyopathy frequently require heart transplantation. Genetic as well as non-genetic factors contribute to restrictive cardiomyopathy and a significant portion of cases are of unknown etiology. However, the genetic forms of restrictive cardiomyopathy and the involved molecular pathomechanisms are only partially understood. In this review, we summarize the current knowledge about primary genetic restrictive cardiomyopathy and describe its genetic landscape, which might be of interest for geneticists as well as for cardiologists.
Collapse
|
13
|
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: 0.3] [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
|
14
|
Kopylova GV, Berg VY, Kochurova AM, Matyushenko AM, Bershitsky SY, Shchepkin DV. The effects of the tropomyosin cardiomyopathy mutations on the calcium regulation of actin-myosin interaction in the atrium and ventricle differ. Biochem Biophys Res Commun 2021; 588:29-33. [PMID: 34942531 DOI: 10.1016/j.bbrc.2021.12.051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/15/2021] [Indexed: 12/17/2022]
Abstract
The molecular mechanisms of pathogenesis of atrial myopathy associated with hypertrophic (HCM) and dilated (DCM) mutations of sarcomeric proteins are still poorly understood. For this, one needs to investigate the effects of the mutations on actin-myosin interaction in the atria separately from ventricles. We compared the impact of the HCM and DCM mutations of tropomyosin (Tpm) on the calcium regulation of the thin filament interaction with atrial and ventricular myosin using an in vitro motility assay. We found that the mutations differently affect the calcium regulation of actin-myosin interaction in the atria and ventricles. The DCM E40K Tpm mutation significantly reduced the maximum sliding velocity of thin filaments with ventricular myosin and its Ca2+-sensitivity. With atrial myosin, its effects were less pronounced. The HCM I172T mutation reduced the Ca2+-sensitivity of the sliding velocity of filaments with ventricular myosin but increased it with the atrial one. The HCM L185R mutation did not affect actin-myosin interaction in the atria. The results indicate that the difference in the effects of Tpm mutations on the actin-myosin interaction in the atria and ventricles may be responsible for the difference in pathological changes in the atrial and ventricular myocardium.
Collapse
Affiliation(s)
- Galina V Kopylova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia
| | - Valentina Y Berg
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia
| | - Anastasia M Kochurova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia
| | - Alexander M Matyushenko
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Sergey Y Bershitsky
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia
| | - Daniil V Shchepkin
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia.
| |
Collapse
|
15
|
Kuruba B, Kaczmarek M, Kęsik-Brodacka M, Fojutowska M, Śliwinska M, Kostyukova AS, Moraczewska J. Structural Effects of Disease-Related Mutations in Actin-Binding Period 3 of Tropomyosin. Molecules 2021; 26:6980. [PMID: 34834072 PMCID: PMC8622905 DOI: 10.3390/molecules26226980] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022] Open
Abstract
Tropomyosin (Tpm) is an actin-binding coiled-coil protein. In muscle, it regulates contractions in a troponin/Ca2+-dependent manner and controls the thin filament lengths at the pointed end. Due to its size and periodic structure, it is difficult to observe small local structural changes in the coiled coil caused by disease-related mutations. In this study, we designed 97-residue peptides, Tpm1.164-154 and Tpm3.1265-155, focusing on the actin-binding period 3 of two muscle isoforms. Using these peptides, we evaluated the effects of cardiomyopathy mutations: I92T and V95A in Tpm1.1, and congenital myopathy mutations R91P and R91C in Tpm3.12. We introduced a cysteine at the N-terminus of each fragment to promote the formation of the coiled-coil structure by disulfide bonds. Dimerization of the designed peptides was confirmed by gel electrophoresis in the presence and absence of dithiothreitol. Using circular dichroism, we showed that all mutations decreased coiled coil stability, with Tpm3.1265-155R91P and Tpm1.164-154I92T having the most drastic effects. Our experiments also indicated that adding the N-terminal cysteine increased coiled coil stability demonstrating that our design can serve as an effective tool in studying the coiled-coil fragments of various proteins.
Collapse
Affiliation(s)
- Balaganesh Kuruba
- Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99163, USA; (B.K.); (A.S.K.)
| | - Marta Kaczmarek
- Department of Biochemistry and Cell Biology, Faculty of Biological Sciences, Kazimierz Wielki University, 85-671 Bydgoszcz, Poland; (M.K.); (M.F.); (M.Ś.)
| | | | - Magdalena Fojutowska
- Department of Biochemistry and Cell Biology, Faculty of Biological Sciences, Kazimierz Wielki University, 85-671 Bydgoszcz, Poland; (M.K.); (M.F.); (M.Ś.)
| | - Małgorzata Śliwinska
- Department of Biochemistry and Cell Biology, Faculty of Biological Sciences, Kazimierz Wielki University, 85-671 Bydgoszcz, Poland; (M.K.); (M.F.); (M.Ś.)
| | - Alla S. Kostyukova
- Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99163, USA; (B.K.); (A.S.K.)
| | - Joanna Moraczewska
- Department of Biochemistry and Cell Biology, Faculty of Biological Sciences, Kazimierz Wielki University, 85-671 Bydgoszcz, Poland; (M.K.); (M.F.); (M.Ś.)
| |
Collapse
|
16
|
James JK, Nanda V. A Folding Insulator Defines Cryptic Domains in Tropomyosin. J Mol Biol 2021; 433:167281. [PMID: 34606830 DOI: 10.1016/j.jmb.2021.167281] [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: 04/06/2021] [Revised: 09/20/2021] [Accepted: 09/27/2021] [Indexed: 10/20/2022]
Abstract
Multidomain proteins are the product of evolutionary selection for diversity of function through concatenation and repurposing of existing modular units of structures. In structures of proteins with multiple domains, components are often globular units stitched together with flexible linkers. Multidomain proteins often fold as multiple distinct order-disorder transitions. However, the relationship between structure and folding is not always straightforward. Tropomyosin binds to actin in muscle and cytoskeletal filaments. The structure is that of a continuous ɑ-helix lacking domain boundaries, but unfolding shows distinct transitions suggesting at least three possible domains do exist. To explore how domains might occur in a continuous structure, we used Lifson-Roig helix-coil models with sequence domains of varying helical nucleation propensities. Of these models, ones with a central folding insulator, separating folding of N- and C-terminal domains, are most consistent with experimental folding studies. The positions of domain boundaries are identified by hydrogen-deuterium exchange mass spectrometry. The presence of structurally cryptic folding domains in tropomyosin could relate to its evolution and explain the uneven distribution of deleterious mutations that lead to various cardiomyopathies.
Collapse
Affiliation(s)
- Jose K James
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | - Vikas Nanda
- Center for Advanced Biotechnology and Medicine and the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, NJ 08854, USA.
| |
Collapse
|
17
|
Naarmann-de Vries IS, Eschenbach J, Dieterich C. Improved nanopore direct RNA sequencing of cardiac myocyte samples by selective mt-RNA depletion. J Mol Cell Cardiol 2021; 163:175-186. [PMID: 34742715 DOI: 10.1016/j.yjmcc.2021.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 01/28/2023]
Abstract
RNA sequencing is a powerful tool to analyze gene expression transcriptome wide. However, RNA sequencing in general and especially the recently developed methods of long read RNA sequencing are still low-throughput and cost-intensive. Here, one important design choice is to concentrate the sequencing capacity on specific parts of the transcriptome. Especially, abundant transcripts as ribosomal RNAs may dominate the available sequencing space, if not removed prior to sequencing. Several methods exist to reduce ribosomal RNA read numbers: either based on enrichment of the relevant fraction (polyA+ RNA) or depletion, respectively degradation of ribosomal RNAs. Furthermore, commercial kits are available to deplete globin transcripts from blood samples. However, so far, no solution exists to deal with other tissue-specific highly abundant transcripts. This is especially of interest in the heart and other muscle derived samples, where reads originating from mitochondrial RNAs make up to 30% of reads in polyA+ selected libraries and around 70% in single cell sequencing experiments. We present a simple method to diminish sequencing of mitochondrial RNAs in Oxford Nanopore direct RNA sequencing libraries by RNase H based clipping of the polyA tail. We show that mt-clipping enables enhanced detection of cytoplasmic mRNAs, among them genes involved in heart development and pathogenesis. Mt-clipping may be applied as well to other sequencing protocols that are based on oligo(dT) priming and can be easily adapted to other tissue-specific high-abundant transcripts.
Collapse
Affiliation(s)
- Isabel S Naarmann-de Vries
- Klaus Tschira Institute for Integrative Computational Cardiology, University Hospital Heidelberg, Germany; Department of Internal Medicine III, University Hospital Heidelberg, Germany; German Center for Cardiovascular Research (DZHK), Partner site Heidelberg/Mannheim, Germany.
| | - Jessica Eschenbach
- Klaus Tschira Institute for Integrative Computational Cardiology, University Hospital Heidelberg, Germany; Department of Internal Medicine III, University Hospital Heidelberg, Germany
| | - Christoph Dieterich
- Klaus Tschira Institute for Integrative Computational Cardiology, University Hospital Heidelberg, Germany; Department of Internal Medicine III, University Hospital Heidelberg, Germany; German Center for Cardiovascular Research (DZHK), Partner site Heidelberg/Mannheim, Germany.
| |
Collapse
|
18
|
Hassoun R, Budde H, Mügge A, Hamdani N. Cardiomyocyte Dysfunction in Inherited Cardiomyopathies. Int J Mol Sci 2021; 22:11154. [PMID: 34681814 PMCID: PMC8541428 DOI: 10.3390/ijms222011154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 01/10/2023] Open
Abstract
Inherited cardiomyopathies form a heterogenous group of disorders that affect the structure and function of the heart. Defects in the genes encoding sarcomeric proteins are associated with various perturbations that induce contractile dysfunction and promote disease development. In this review we aimed to outline the functional consequences of the major inherited cardiomyopathies in terms of myocardial contraction and kinetics, and to highlight the structural and functional alterations in some sarcomeric variants that have been demonstrated to be involved in the pathogenesis of the inherited cardiomyopathies. A particular focus was made on mutation-induced alterations in cardiomyocyte mechanics. Since no disease-specific treatments for familial cardiomyopathies exist, several novel agents have been developed to modulate sarcomere contractility. Understanding the molecular basis of the disease opens new avenues for the development of new therapies. Furthermore, the earlier the awareness of the genetic defect, the better the clinical prognostication would be for patients and the better the prevention of development of the disease.
Collapse
Affiliation(s)
- Roua Hassoun
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Heidi Budde
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| |
Collapse
|
19
|
Aziz A, Musiol SK, Moody WE, Pickup L, Cooper R, Lip GYH. Clinical prediction of genotypes in hypertrophic cardiomyopathy: A systematic review. Eur J Clin Invest 2021; 51:e13593. [PMID: 33948946 DOI: 10.1111/eci.13593] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/14/2021] [Accepted: 03/18/2021] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac condition and the most common cause of sudden cardiac death (SCD) in patients below the age of 35. Genetic testing is a vital part of HCM diagnostics, yet correlation with clinical phenotypes remains complex. Identifying clinical predictors of informative genetic testing may prevent unnecessary investigations and improve cost-effectiveness of services. This article reviews the current literature pertinent to identifying such predictors. METHODS Five literature databases were screened using a suitably designed search strategy. Studies investigating the correlation between having a positive genetic test for HCM and a range of clinical and radiological parameters were included in the systematic review. RESULTS Twenty-nine observational studies of a total of 9,486 patients were included. The main predictors of informative genetic testing were younger age, higher septal thickness, reverse septal curvature, family history of HCM and SCD and the absence of hypertension. Two externally validated scoring systems have also been developed: the Mayo and Toronto scores. Novel imaging markers and complex algorithmic models are emerging predictors. CONCLUSION Using clinical predictors to decide whom to test is a feasible alternative to investigating all comers. Nonetheless, currently there is not enough evidence to unequivocally recommend for or against this strategy. Further validation of current predictors and identification of new ones remain open research avenues.
Collapse
Affiliation(s)
- Amir Aziz
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | | | - William E Moody
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Luke Pickup
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Rob Cooper
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK.,Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
20
|
Sewanan LR, Park J, Rynkiewicz MJ, Racca AW, Papoutsidakis N, Schwan J, Jacoby DL, Moore JR, Lehman W, Qyang Y, Campbell SG. Loss of crossbridge inhibition drives pathological cardiac hypertrophy in patients harboring the TPM1 E192K mutation. J Gen Physiol 2021; 153:212516. [PMID: 34319370 PMCID: PMC8321830 DOI: 10.1085/jgp.202012640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/14/2021] [Accepted: 07/09/2021] [Indexed: 01/10/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is an inherited disorder caused primarily by mutations to thick and thinfilament proteins. Although thin filament mutations are less prevalent than their oft-studied thick filament counterparts, they are frequently associated with severe patient phenotypes and can offer important insight into fundamental disease mechanisms. We have performed a detailed study of tropomyosin (TPM1) E192K, a variant of uncertain significance associated with HCM. Molecular dynamics revealed that E192K results in a more flexible TPM1 molecule, which could affect its ability to regulate crossbridges. In vitro motility assays of regulated actin filaments containing TPM1 E192K showed an overall loss of Ca2+ sensitivity. To understand these effects, we used multiscale computational models that suggested a subtle phenotype in which E192K leads to an inability to completely inhibit actin-myosin crossbridge activity at low Ca2+. To assess the physiological impact of the mutation, we generated patient-derived engineered heart tissues expressing E192K. These tissues showed disease features similar to those of the patients, including cellular hypertrophy, hypercontractility, and diastolic dysfunction. We hypothesized that excess residual crossbridge activity could be triggering cellular hypertrophy, even if the overall Ca2+ sensitivity was reduced by E192K. To test this hypothesis, the cardiac myosin-specific inhibitor mavacamten was applied to patient-derived engineered heart tissues for 4 d followed by 24 h of washout. Chronic mavacamten treatment abolished contractile differences between control and TPM1 E192K engineered heart tissues and reversed hypertrophy in cardiomyocytes. These results suggest that the TPM1 E192K mutation triggers cardiomyocyte hypertrophy by permitting excess residual crossbridge activity. These studies also provide direct evidence that myosin inhibition by mavacamten can counteract the hypertrophic effects of mutant tropomyosin.
Collapse
Affiliation(s)
- Lorenzo R Sewanan
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Jinkyu Park
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT.,Yale Stem Cell Center, Yale School of Medicine, New Haven, CT
| | - Michael J Rynkiewicz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA
| | - Alice W Racca
- Department of Biological Sciences, University of Massachusetts, Lowell, MA
| | - Nikolaos Papoutsidakis
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
| | - Jonas Schwan
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Daniel L Jacoby
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
| | - Jeffrey R Moore
- Department of Biological Sciences, University of Massachusetts, Lowell, MA
| | - William Lehman
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA
| | - Yibing Qyang
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT.,Yale Stem Cell Center, Yale School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program, Yale University, New Haven, CT.,Department of Pathology, Yale University, New Haven, CT
| | - Stuart G Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT.,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT
| |
Collapse
|
21
|
Cao J, Routh AL, Kuyumcu-Martinez MN. Nanopore sequencing reveals full-length Tropomyosin 1 isoforms and their regulation by RNA-binding proteins during rat heart development. J Cell Mol Med 2021; 25:8352-8362. [PMID: 34302435 PMCID: PMC8419188 DOI: 10.1111/jcmm.16795] [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: 05/04/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 12/11/2022] Open
Abstract
Alternative splicing (AS) contributes to the diversity of the proteome by producing multiple isoforms from a single gene. Although short‐read RNA‐sequencing methods have been the gold standard for determining AS patterns of genes, they have a difficulty in defining full‐length mRNA isoforms assembled using different exon combinations. Tropomyosin 1 (TPM1) is an actin‐binding protein required for cytoskeletal functions in non‐muscle cells and for contraction in muscle cells. Tpm1 undergoes AS regulation to generate muscle versus non‐muscle TPM1 protein isoforms with distinct physiological functions. It is unclear which full‐length Tpm1 isoforms are produced via AS and how they are regulated during heart development. To address these, we utilized nanopore long‐read cDNA sequencing without gene‐specific PCR amplification. In rat hearts, we identified full‐length Tpm1 isoforms composed of distinct exons with specific exon linkages. We showed that Tpm1 undergoes AS transitions during embryonic heart development such that muscle‐specific exons are connected generating predominantly muscle‐specific Tpm1 isoforms in adult hearts. We found that the RNA‐binding protein RBFOX2 controls AS of rat Tpm1 exon 6a, which is important for cooperative actin binding. Furthermore, RBFOX2 regulates Tpm1 AS of exon 6a antagonistically to the RNA‐binding protein PTBP1. In sum, we defined full‐length Tpm1 isoforms with different exon combinations that are tightly regulated during cardiac development and provided insights into the regulation of Tpm1 AS by RNA‐binding proteins. Our results demonstrate that nanopore sequencing is an excellent tool to determine full‐length AS variants of muscle‐enriched genes.
Collapse
Affiliation(s)
- Jun Cao
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Andrew L Routh
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA.,Sealy Centre for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Muge N Kuyumcu-Martinez
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA.,Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, USA.,Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
22
|
Chen YJ, Chien CS, Chiang CE, Chen CH, Cheng HM. From Genetic Mutations to Molecular Basis of Heart Failure Treatment: An Overview of the Mechanism and Implication of the Novel Modulators for Cardiac Myosin. Int J Mol Sci 2021; 22:6617. [PMID: 34205587 PMCID: PMC8234187 DOI: 10.3390/ijms22126617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) is a syndrome encompassing several important etiologies that lead to the imbalance between oxygen demand and supply. Despite the usage of guideline-directed medical therapy for HF has shown better outcomes, novel therapeutic strategies are desirable, especially for patients with preserved or mildly reduced left ventricular ejection fraction. In this regard, understanding the molecular basis for cardiomyopathies is expected to fill in the knowledge gap and generate new therapies to improve prognosis for HF. This review discusses an evolutionary mechanism designed to regulate cardiac contraction and relaxation through the most often genetically determined cardiomyopathies associated with HF. In addition, both the myosin inhibitor and myosin activator are promising new treatments for cardiomyopathies. A comprehensive review from genetic mutations to the molecular basis of direct sarcomere modulators will help shed light on future studies for a better characterization of HF etiologies and potential therapeutic targets.
Collapse
Affiliation(s)
- Yu-Jen Chen
- Department of Internal Medicine, Division of Cardiovascular Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan;
- Department of Internal Medicine, Division of Cardiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Chian-Shiu Chien
- Innovative Cellular Therapy Center, Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
| | - Chern-En Chiang
- General Clinical Research Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
- Department of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei 112304, Taiwan
| | - Chen-Huan Chen
- Department of Medical Education, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
- College of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
| | - Hao-Min Cheng
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei 112304, Taiwan
- Center for Evidence-Based Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| |
Collapse
|
23
|
Wu H, Jiang W, Ji G, Xu R, Zhou G, Yu H. Exploring microRNA target genes and identifying hub genes in bladder cancer based on bioinformatic analysis. BMC Urol 2021; 21:90. [PMID: 34112125 PMCID: PMC8194198 DOI: 10.1186/s12894-021-00857-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/04/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Bladder cancer (BC) is the second most frequent malignancy of the urinary system. The aim of this study was to identify key microRNAs (miRNAs) and hub genes associated with BC as well as analyse their targeted relationships. METHODS According to the microRNA dataset GSE112264 and gene microarray dataset GSE52519, differentially expressed microRNAs (DEMs) and differentially expressed genes (DEGs) were obtained using the R limma software package. The FunRich software database was used to predict the miRNA-targeted genes. The overlapping common genes (OCGs) between miRNA-targeted genes and DEGs were screened to construct the PPI network. Then, gene ontology (GO) analysis was performed through the "cluster Profiler" and "org.Hs.eg.db" R packages. The differential expression analysis and hierarchical clustering of these hub genes were analysed through the GEPIA and UCSC Cancer Genomics Browser databases, respectively. KEGG pathway enrichment analyses of hub genes were performed through gene set enrichment analysis (GSEA). RESULTS A total of 12 DEMs and 10 hub genes were identified. Differential expression analysis of the hub genes using the GEPIA database was consistent with the results for the UCSC Cancer Genomics Browser database. The results indicated that these hub genes were oncogenes, but VCL, TPM2, and TPM1 were tumour suppressor genes. The GSEA also showed that hub genes were most enriched in those pathways that were closely associated with tumour proliferation and apoptosis. CONCLUSIONS In this study, we built a miRNA-mRNA regulatory targeted network, which explores an understanding of the pathogenesis of cancer development and provides key evidence for novel targeted treatments for BC.
Collapse
Affiliation(s)
- Hongjian Wu
- Department of Urology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, 317000, Zhejiang, People's Republic of China
| | - Wubing Jiang
- Department of Urology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, 317000, Zhejiang, People's Republic of China
| | - Guanghua Ji
- Department of Urology, Taizhou Municipal Hospital, Taizhou, 317000, Zhejiang, People's Republic of China
| | - Rong Xu
- Department of Urology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, 317000, Zhejiang, People's Republic of China
| | - Gaobo Zhou
- Department of Urology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, 317000, Zhejiang, People's Republic of China
| | - Hongyuan Yu
- Department of Urology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, 317000, Zhejiang, People's Republic of China.
| |
Collapse
|
24
|
Mutations Q93H and E97K in TPM2 Disrupt Ca-Dependent Regulation of Actin Filaments. Int J Mol Sci 2021; 22:ijms22084036. [PMID: 33919826 PMCID: PMC8070786 DOI: 10.3390/ijms22084036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Tropomyosin is a two-chain coiled coil protein, which together with the troponin complex controls interactions of actin with myosin in a Ca2+-dependent manner. In fast skeletal muscle, the contractile actin filaments are regulated by tropomyosin isoforms Tpm1.1 and Tpm2.2, which form homo- and heterodimers. Mutations in the TPM2 gene encoding isoform Tpm2.2 are linked to distal arthrogryposis and congenital myopathy-skeletal muscle diseases characterized by hyper- and hypocontractile phenotypes, respectively. In this work, in vitro functional assays were used to elucidate the molecular mechanisms of mutations Q93H and E97K in TPM2. Both mutations tended to decrease actin affinity of homo-and heterodimers in the absence and presence of troponin and Ca2+, although the effect of Q93H was stronger. Changes in susceptibility of tropomyosin to trypsin digestion suggested that the mutations diversified dynamics of tropomyosin homo- and heterodimers on the filament. The presence of Q93H in homo- and heterodimers strongly decreased activation of the actomyosin ATPase and reduced sensitivity of the thin filament to [Ca2+]. In contrast, the presence of E97K caused hyperactivation of the ATPase and increased sensitivity to [Ca2+]. In conclusion, the hypo- and hypercontractile phenotypes associated with mutations Q93H and E97K in Tpm2.2 are caused by defects in Ca2+-dependent regulation of actin-myosin interactions.
Collapse
|
25
|
Pham AK, Miller M, Rosenthal P, Das S, Weng N, Jang S, Kurten RC, Badrani J, Doherty TA, Oliver B, Broide DH. ORMDL3 expression in ASM regulates hypertrophy, hyperplasia via TPM1 and TPM4, and contractility. JCI Insight 2021; 6:136911. [PMID: 33661765 PMCID: PMC8119187 DOI: 10.1172/jci.insight.136911] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/26/2021] [Indexed: 12/21/2022] Open
Abstract
ORM1-like 3 (ORMDL3) has strong genetic linkage to childhood onset asthma. To determine whether ORMDL3 selective expression in airway smooth muscle (ASM) influences ASM function, we used Cre-loxP techniques to generate transgenic mice (hORMDL3Myh11eGFP-cre), which express human ORMDL3 selectively in smooth muscle cells. In vitro studies of ASM cells isolated from the bronchi of hORMDL3Myh11eGFP-cre mice demonstrated that they developed hypertrophy (quantitated by FACS and image analysis), developed hyperplasia (assessed by BrdU incorporation), and expressed increased levels of tropomysin proteins TPM1 and TPM4. siRNA knockdown of TPM1 or TPM4 demonstrated their importance to ORMDL3-mediated ASM proliferation but not hypertrophy. In addition, ASM derived from hORMDL3Myh11eGFP-cre mice had increased contractility to histamine in vitro, which was associated with increased levels of intracellular Ca2+; increased cell surface membrane Orai1 Ca2+ channels, which mediate influx of Ca2+ into the cytoplasm; and increased expression of ASM contractile genes sarco/endoplasmic reticulum Ca2+ ATPase 2b and smooth muscle 22. In vivo studies of hORMDL3Myh11eGFP-cre mice demonstrated that they had a spontaneous increase in ASM and airway hyperreactivity (AHR). ORMDL3 expression in ASM thus induces changes in ASM (hypertrophy, hyperplasia, increased contractility), which may explain the contribution of ORMDL3 to the development of AHR in childhood onset asthma, which is highly linked to ORMDL3 on chromosome 17q12-21.
Collapse
Affiliation(s)
- Alexa K. Pham
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Marina Miller
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Peter Rosenthal
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Sudipta Das
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Ning Weng
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Sunghoon Jang
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Richard C. Kurten
- Department of Pediatrics, Arkansas Children’s Research Institute, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jana Badrani
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Taylor A. Doherty
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Veterans Affairs San Diego Health Care System, La Jolla, California, USA
| | - Brian Oliver
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - David H. Broide
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| |
Collapse
|
26
|
Racca AW, Rynkiewicz MJ, LaFave N, Ghosh A, Lehman W, Moore JR. M8R tropomyosin mutation disrupts actin binding and filament regulation: The beginning affects the middle and end. J Biol Chem 2020; 295:17128-17137. [PMID: 33020181 PMCID: PMC7863880 DOI: 10.1074/jbc.ra120.014713] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/28/2020] [Indexed: 11/06/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is associated with mutations in cardiomyocyte sarcomeric proteins, including α-tropomyosin. In conjunction with troponin, tropomyosin shifts to regulate actomyosin interactions. Tropomyosin molecules overlap via tropomyosin-tropomyosin head-to-tail associations, forming a continuous strand along the thin filament. These associations are critical for propagation of tropomyosin's reconfiguration along the thin filament and key for the cooperative switching between heart muscle contraction and relaxation. Here, we tested perturbations in tropomyosin structure, biochemistry, and function caused by the DCM-linked mutation, M8R, which is located at the overlap junction. Localized and nonlocalized structural effects of the mutation were found in tropomyosin that ultimately perturb its thin filament regulatory function. Comparison of mutant and WT α-tropomyosin was carried out using in vitro motility assays, CD, actin co-sedimentation, and molecular dynamics simulations. Regulated thin filament velocity measurements showed that the presence of M8R tropomyosin decreased calcium sensitivity and thin filament cooperativity. The co-sedimentation of actin and tropomyosin showed weakening of actin-mutant tropomyosin binding. The binding of troponin T's N terminus to the actin-mutant tropomyosin complex was also weakened. CD and molecular dynamics indicate that the M8R mutation disrupts the four-helix bundle at the head-to-tail junction, leading to weaker tropomyosin-tropomyosin binding and weaker tropomyosin-actin binding. Molecular dynamics revealed that altered end-to-end bond formation has effects extending toward the central region of the tropomyosin molecule, which alter the azimuthal position of tropomyosin, likely disrupting the mutant thin filament response to calcium. These results demonstrate that mutation-induced alterations in tropomyosin-thin filament interactions underlie the altered regulatory phenotype and ultimately the pathogenesis of DCM.
Collapse
Affiliation(s)
- Alice Ward Racca
- Department of Biological Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts, USA
| | - Michael J Rynkiewicz
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Nicholas LaFave
- Department of Biological Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts, USA
| | - Anita Ghosh
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, Massachusetts, USA
| | - William Lehman
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jeffrey R Moore
- Department of Biological Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts, USA.
| |
Collapse
|
27
|
Nefedova VV, Koubassova NA, Borzova VA, Kleymenov SY, Tsaturyan AK, Matyushenko AM, Levitsky DI. Tropomyosin pseudo-phosphorylation can rescue the effects of cardiomyopathy-associated mutations. Int J Biol Macromol 2020; 166:424-434. [PMID: 33129908 DOI: 10.1016/j.ijbiomac.2020.10.201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/28/2022]
Abstract
We applied various methods to investigate how mutations S283D and S61D that mimic phosphorylation of tropomyosin (Tpm) affect structural and functional properties of cardiac Tpm carrying cardiomyopathy-associated mutations in different parts of its molecule. Using differential scanning calorimetry and molecular dynamics, we have shown that the S61D mutation (but not the S283 mutation) causes significant destabilization of the N-terminal part of the Tpm molecule independently of the absence or presence of cardiomyopathy-associated mutations. Our results obtained by cosedimentation of Tpm with F-actin demonstrated that both S283D and S61D mutations can reduce or even eliminate undesirable changes in Tpm affinity for F-actin caused by some cardiomyopathy-associated mutations. The results indicate that Tpm pseudo-phosphorylation by mutations S283D or S61D can rescue the effects of mutations in the TPM1 gene encoding a cardiac isoform of Tpm that lead to the development of such severe inherited heart diseases as hypertrophic or dilated cardiomyopathies.
Collapse
Affiliation(s)
- Victoria V Nefedova
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky prosp. 33, 119071 Moscow, Russia
| | - Natalia A Koubassova
- Institute of Mechanics, Moscow State University, Mitchurinsky prosp. 1, 119192 Moscow, Russia
| | - Vera A Borzova
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky prosp. 33, 119071 Moscow, Russia
| | - Sergey Y Kleymenov
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky prosp. 33, 119071 Moscow, Russia; Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, 119334 Moscow, Russia
| | - Andrey K Tsaturyan
- Institute of Mechanics, Moscow State University, Mitchurinsky prosp. 1, 119192 Moscow, Russia
| | - Alexander M Matyushenko
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky prosp. 33, 119071 Moscow, Russia
| | - Dmitrii I Levitsky
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky prosp. 33, 119071 Moscow, Russia.
| |
Collapse
|
28
|
Looking for Targets to Restore the Contractile Function in Congenital Myopathy Caused by Gln 147Pro Tropomyosin. Int J Mol Sci 2020; 21:ijms21207590. [PMID: 33066566 PMCID: PMC7589864 DOI: 10.3390/ijms21207590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/06/2020] [Accepted: 10/11/2020] [Indexed: 12/15/2022] Open
Abstract
We have used the technique of polarized microfluorimetry to obtain new insight into the pathogenesis of skeletal muscle disease caused by the Gln147Pro substitution in β-tropomyosin (Tpm2.2). The spatial rearrangements of actin, myosin and tropomyosin in the single muscle fiber containing reconstituted thin filaments were studied during simulation of several stages of ATP hydrolysis cycle. The angular orientation of the fluorescence probes bound to tropomyosin was found to be changed by the substitution and was characteristic for a shift of tropomyosin strands closer to the inner actin domains. It was observed both in the absence and in the presence of troponin, Ca2+ and myosin heads at all simulated stages of the ATPase cycle. The mutant showed higher flexibility. Moreover, the Gln147Pro substitution disrupted the myosin-induced displacement of tropomyosin over actin. The irregular positioning of the mutant tropomyosin caused premature activation of actin monomers and a tendency to increase the number of myosin cross-bridges in a state of strong binding with actin at low Ca2+.
Collapse
|
29
|
Dorsch LM, Kuster DWD, Jongbloed JDH, Boven LG, van Spaendonck-Zwarts KY, Suurmeijer AJH, Vink A, du Marchie Sarvaas GJ, van den Berg MP, van der Velden J, Brundel BJJM, van der Zwaag PA. The effect of tropomyosin variants on cardiomyocyte function and structure that underlie different clinical cardiomyopathy phenotypes. Int J Cardiol 2020; 323:251-258. [PMID: 32882290 DOI: 10.1016/j.ijcard.2020.08.101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/20/2020] [Accepted: 08/28/2020] [Indexed: 12/27/2022]
Abstract
Background - Variants within the alpha-tropomyosin gene (TPM1) cause dominantly inherited cardiomyopathies, including dilated (DCM), hypertrophic (HCM) and restrictive (RCM) cardiomyopathy. Here we investigated whether TPM1 variants observed in DCM and HCM patients affect cardiomyocyte physiology differently. Methods - We identified a large family with DCM carrying a recently identified TPM1 gene variant (T201M) and a child with RCM with compound heterozygote TPM1 variants (E62Q and M281T) whose family members carrying single variants show diastolic dysfunction and HCM. The effects of TPM1 variants (T201M, E62Q or M281T) and of a plasmid containing both the E62Q and M281T variants on single-cell Ca2+ transients (CaT) in HL-1 cardiomyocytes were studied. To define toxic threshold levels, we performed dose-dependent transfection of TPM1 variants. In addition, cardiomyocyte structure was studied in human cardiac biopsies with TPM1 variants. Results - Overexpression of TPM1 variants led to time-dependent progressive deterioration of CaT, with the smallest effect seen for E62Q and larger and similar effects seen for the T201M and M281T variants. Overexpression of E62Q/M281T did not exacerbate the effects seen with overexpression of a single TPM1 variant. T201M (DCM) replaced endogenous tropomyosin dose-dependently, while M281T (HCM) did not. Human cardiac biopsies with TPM1 variants revealed loss of sarcomeric structures. Conclusion - All TPM1 variants result in reduced cardiomyocyte CaT amplitudes and loss of sarcomeric structures. These effects may underlie pathophysiology of different cardiomyopathy phenotypes.
Collapse
Affiliation(s)
- Larissa M Dorsch
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
| | - Diederik W D Kuster
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Jan D H Jongbloed
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ludolf G Boven
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Karin Y van Spaendonck-Zwarts
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Albert J H Suurmeijer
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Maarten P van den Berg
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Paul A van der Zwaag
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
30
|
Matyushenko AM, Levitsky DI. Molecular Mechanisms of Pathologies of Skeletal and Cardiac Muscles Caused by Point Mutations in the Tropomyosin Genes. BIOCHEMISTRY (MOSCOW) 2020; 85:S20-S33. [PMID: 32087052 DOI: 10.1134/s0006297920140023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The review is devoted to tropomyosin (Tpm) - actin-binding protein, which plays a crucial role in the regulation of contraction of skeletal and cardiac muscles. Special attention is paid to myopathies and cardiomyopathies - severe hereditary diseases of skeletal and cardiac muscles associated with point mutations in Tpm genes. The current views on the molecular mechanisms of these diseases and the effects of such mutations on the Tpm structure and functions are considered in detail. Besides, some part of the review is devoted to analysis of the properties of Tpm homodimers and heterodimers with myopathic substitutions of amino acid residues in only one of the two chains of the Tpm dimeric molecule.
Collapse
Affiliation(s)
- A M Matyushenko
- Bach Institute of Biochemistry, Federal Research Center on Fundamentals of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia.
| | - D I Levitsky
- Bach Institute of Biochemistry, Federal Research Center on Fundamentals of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia. .,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
| |
Collapse
|
31
|
Lehman W, Rynkiewicz MJ, Moore JR. A new twist on tropomyosin binding to actin filaments: perspectives on thin filament function, assembly and biomechanics. J Muscle Res Cell Motil 2020; 41:23-38. [PMID: 30771202 PMCID: PMC6697252 DOI: 10.1007/s10974-019-09501-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 02/07/2019] [Indexed: 02/07/2023]
Abstract
Tropomyosin, best known for its role in the steric regulation of muscle contraction, polymerizes head-to-tail to form cables localized along the length of both muscle and non-muscle actin-based thin filaments. In skeletal and cardiac muscles, tropomyosin, under the control of troponin and myosin, moves in a cooperative manner between blocked, closed and open positions on filaments, thereby masking and exposing actin-binding sites necessary for myosin crossbridge head interactions. While the coiled-coil signature of tropomyosin appears to be simple, closer inspection reveals surprising structural complexity required to perform its role in steric regulation. For example, component α-helices of coiled coils are typically zippered together along a continuous core hydrophobic stripe. Tropomyosin, however, contains a number of anomalous, functionally controversial, core amino acid residues. We argue that the atypical residues at this interface, including clusters of alanines and a charged aspartate, are required for preshaping tropomyosin to readily fit to the surface of the actin filament, but do so without compromising tropomyosin rigidity once the filament is assembled. Indeed, persistence length measurements of tropomyosin are characteristic of a semi-rigid cable, in this case conducive to cooperative movement on thin filaments. In addition, we also maintain that tropomyosin displays largely unrecognized and residue-specific torsional variance, which is involved in optimizing contacts between actin and tropomyosin on the assembled thin filament. Corresponding twist-induced stiffness may also enhance cooperative translocation of tropomyosin across actin filaments. We conclude that anomalous core residues of tropomyosin facilitate thin filament regulatory behavior in a multifaceted way.
Collapse
Affiliation(s)
- William Lehman
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, Massachusetts, U.S.A
| | - Michael J. Rynkiewicz
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, Massachusetts, U.S.A
| | - Jeffrey R. Moore
- Department of Biological Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts, U.S.A
| |
Collapse
|
32
|
Borovikov YS, Karpicheva OE, Avrova SV, Simonyan AO, Sirenko VV, Redwood CS. The molecular mechanism of muscle dysfunction associated with the R133W mutation in Tpm2.2. Biochem Biophys Res Commun 2019; 523:258-262. [PMID: 31864708 DOI: 10.1016/j.bbrc.2019.12.061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 12/13/2019] [Indexed: 12/16/2022]
Abstract
Ghost muscle fibres reconstituted with myosin heads labeled with the fluorescent probe 1,5-IAEDANS were used for analysis of muscle fibre dysfunction associated with the R133W mutation in β-tropomyosin (Tpm2.2). By using polarized microscopy, we showed that at high Ca2+ the R133W mutation in both αβ-Tpm heterodimers and ββ-Tpm homodimers decreases the amount of the myosin heads strongly bound to F-actin and the number of switched-on actin monomers, with this effect being stronger for ββ-Tpm. This mutation also inhibits the shifting of the R133W-Tpm strands towards the open position and the efficiency of the cross-bridge work. At low Ca2+, the amount of the strongly bound myosin heads is lower for R133W-Tpms than for WT-Tpms which may contribute to a low myofilament Ca2+-sensitivity of the R133W-Tpms. It is concluded that freezing of the mutant αβ- or ββ-Tpm close to the blocked position inhibits the strong binding of the cross-bridges and the switching on of actin monomers which may be the reason for muscle weakness associated with the R133W mutation in β-tropomyosin. The use of reagents that activate myosin may be appropriate to restore muscle function in patients with the R133W mutation.
Collapse
Affiliation(s)
- Yurii S Borovikov
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Av., St. Petersburg, 194064, Russia.
| | - Olga E Karpicheva
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Av., St. Petersburg, 194064, Russia
| | - Stanislava V Avrova
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Av., St. Petersburg, 194064, Russia
| | - Armen O Simonyan
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Av., St. Petersburg, 194064, Russia
| | - Vladimir V Sirenko
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Av., St. Petersburg, 194064, Russia
| | - Charles S Redwood
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| |
Collapse
|
33
|
Aboelkassem Y, McCabe KJ, Huber GA, Regnier M, McCammon JA, McCulloch AD. A Stochastic Multiscale Model of Cardiac Thin Filament Activation Using Brownian-Langevin Dynamics. Biophys J 2019; 117:2255-2272. [PMID: 31547973 PMCID: PMC6990154 DOI: 10.1016/j.bpj.2019.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 11/16/2022] Open
Abstract
We use Brownian-Langevin dynamics principles to derive a coarse-graining multiscale myofilament model that can describe the thin-filament activation process during contraction. The model links atomistic molecular simulations of protein-protein interactions in the thin-filament regulatory unit to sarcomere-level activation dynamics. We first calculate the molecular interaction energy between tropomyosin and actin surface using Brownian dynamics simulations. This energy profile is then generalized to account for the observed tropomyosin transitions between its regulatory stable states. The generalized energy landscape then served as a basis for developing a filament-scale model using Langevin dynamics. This integrated analysis, spanning molecular to thin-filament scales, is capable of tracking the events of the tropomyosin conformational changes as it moves over the actin surface. The tropomyosin coil with flexible overlap regions between adjacent tropomyosins is represented in the model as a system of coupled stochastic ordinary differential equations. The proposed multiscale approach provides a more detailed molecular connection between tropomyosin dynamics, the trompomyosin-actin interaction-energy landscape, and the generated force by the sarcomere.
Collapse
Affiliation(s)
- Yasser Aboelkassem
- Department of Bioengineering, University of California San Diego, La Jolla, California.
| | - Kimberly J McCabe
- Department of Bioengineering, University of California San Diego, La Jolla, California
| | - Gary A Huber
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, Washington
| | - J Andrew McCammon
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, La Jolla, California
| |
Collapse
|
34
|
Sundar S, Rynkiewicz MJ, Ghosh A, Lehman W, Moore JR. Cardiomyopathy Mutation Alters End-to-End Junction of Tropomyosin and Reduces Calcium Sensitivity. Biophys J 2019; 118:303-312. [PMID: 31882250 DOI: 10.1016/j.bpj.2019.11.3396] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/30/2019] [Accepted: 11/18/2019] [Indexed: 12/24/2022] Open
Abstract
Muscle contraction is governed by tropomyosin (Tpm) shifting azimuthally between three states on F-actin (B-, C-, and M-states) in response to calcium binding to troponin and actomyosin cross-bridge formation. The Tpm coiled coil polymerizes head to tail along the long-pitch helix of F-actin to form continuous superhelical cables that wrap around the actin filaments. The end-to-end bonds formed between the N- and C-terminus of adjacent Tpm molecules define Tpm continuity and play a critical role in the ability of Tpm to cooperatively bind to actin, thus facilitating Tpm conformational switching to cooperatively propagate along F-actin. We expect that a missense mutation in this critical overlap region associated with dilated cardiomyopathy, A277V, will alter Tpm binding and thin filament activation by altering the overlap structure. Here, we used cosedimentation assays and in vitro motility assays to determine how the mutation alters Tpm binding to actin and its ability to regulate actomyosin interactions. Analytical viscometry coupled with molecular dynamics simulations showed that the A277V mutation results in enhanced Tpm end-to-end bond strength and a reduced curvature of the Tpm overlap domain. The mutant Tpm exhibited enhanced actin-Tpm binding affinity, consistent with overlap stabilization. The observed A277V-induced decrease in cooperative activation observed with regulated thin filament motility indicates that increased overlap stabilization is not correlated with Tpm-Tpm overlap binding strength or mechanical rigidity as is often assumed. Instead, A277V-induced structural changes result in local and delocalized increases in Tpm flexibility and prominent coiled-coil twisting in pseudorepeat 4. An A277V-induced decrease in Ca2+ sensitivity, consistent with a mutation-induced bolstering of the B-state Tpm-actin electrostatic contacts and an increased Tpm troponin T1 binding affinity, was also observed.
Collapse
Affiliation(s)
- SaiLavanyaa Sundar
- Department of Biological Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts
| | - Michael J Rynkiewicz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts
| | - Anita Ghosh
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts
| | - William Lehman
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts
| | - Jeffrey R Moore
- Department of Biological Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts.
| |
Collapse
|
35
|
Yao Q, Zhang W, Zhang T. Association of single nucleotide polymorphisms in the 3'UTR region of TPM1 gene with dilated cardiomyopathy: A case-control study. Medicine (Baltimore) 2019; 98:e17710. [PMID: 31689804 PMCID: PMC6946328 DOI: 10.1097/md.0000000000017710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Tropomyosin 1 (TPM1) is a protein that constitutes the sarcomere filaments and is encoded by the TPM1 gene. The aim of the present study is to investigate the correlation between the 3' untranslated region (3'UTR) single nucleotide polymorphisms (SNPs) of the TPM1 gene and dilated cardiomyopathy (DCM).A total of 245 patients with DCM and 245 healthy controls were recruited with 5 ml of venous blood. Genomic DNA was extracted to analyze the TPM1 gene rs12148828, rs11558748, rs707602, rs6738, rs7178040 loci genotypes, and the plasma miR-21 level was analyzed by reverse transcription-PCR (RT-PCR).The risk of DCM development in the rs6738 locus G allele carriers were 1.69 times more than A allele carriers (95% CI: 1.22-2.33, P = .001). Age and gender had no effect on the association of TPM1 gene SNPs with DCM risk (P > .05). The plasma miR-21 level of TPM1 gene rs6738 locus AA carriers was significantly higher than that of the AG and GG genotypes (P < .001).The SNPs of TPM1 gene rs6738 locus is associated with the risk of DCM, which may be related to the abnormal increase of miR-21 level in DCM patients, but further research is needed to prove the causal relationship between miR-21 level and DCM risk.
Collapse
Affiliation(s)
- Qiang Yao
- Department of Cardiology, Zhejiang Chinese Medicine and Western Medicine Integrated Hospital/Hangzhou Red Cross Hospital, Hangzhou Zhejiang
| | - Wei Zhang
- Department of Cardiology, Chengwu county people's hospital, Chengwu County, Shandong Province
| | - Tianjie Zhang
- Department of Cardiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
36
|
Cardiomyopathy-associated mutations in tropomyosin differently affect actin–myosin interaction at single-molecule and ensemble levels. J Muscle Res Cell Motil 2019; 40:299-308. [DOI: 10.1007/s10974-019-09560-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 10/15/2019] [Indexed: 01/31/2023]
|
37
|
miRNA‑183‑5p.1 promotes the migration and invasion of gastric cancer AGS cells by targeting TPM1. Oncol Rep 2019; 42:2371-2381. [PMID: 31638242 PMCID: PMC6859460 DOI: 10.3892/or.2019.7354] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 09/12/2019] [Indexed: 01/13/2023] Open
Abstract
MicroRNA-183 (miR-183) is a small, non-coding RNA that is involved in post-transcriptional processes, is upregulated in gastric cancer and acts as an oncogene in cancer migration. Although fragmentary reports have demonstrated the importance of miR-183 in gastric cancer, its biofunctions and regulatory effects are still unknown. In the present study, the gene and protein expression levels were determined by reverse transcription-quantitative PCR and western blot analysis. The connection between miR-183-5p.1 and tropomyosin 1 (TPM1) was tested through luciferase reporter experiments. Cell viability, apoptosis and related proteins were detected by MTT assay, flow cytometry, immunofluorescence and western blotting, respectively. The migration and invasion of AGS cells modulated by miR-183-5p.1 were analyzed by Transwell assay. TPM1 expression was found to be decreased in gastric cancer tissues and cell lines when compared with normal and adjacent tissues and gastric epithelial cells, and was regulated by miR-183-5p.1 targeting TPM1. miR-183-5p.1 overexpression facilitated the growth and suppressed the death of AGS cells through Bcl-2 and P53 proteins. In addition, miR-183-5p.1 restricted TPM1, TPM2 and TPM3 protein expression in AGS cells. The excessive levels of miR-183-5p.1 promoted the migration and invasion of AGS cells, and inhibited the apoptosis of AGS cells. However, the knockdown of miR-183-5p.1 induced the opposite in AGS cells. In conclusion, miR-183-5p.1 promotes cell proliferation, migration and invasion by downregulating TPM1 and deactivating the Bcl-2/P53 signaling pathways in gastric cancer, indicating that miR-183-5p.1 and TPM1 may be potential targets for the diagnosis or therapy of gastric cancer in the future.
Collapse
|
38
|
Aboelkassem Y, Powers JD, McCabe KJ, McCulloch AD. Multiscale Models of Cardiac Muscle Biophysics and Tissue Remodeling in Hypertrophic Cardiomyopathies. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019; 11:35-44. [PMID: 31886450 DOI: 10.1016/j.cobme.2019.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Myocardial hypertrophy is the result of sustained perturbations to the mechanical and/or neurohormonal homeostasis of cardiac cells and is driven by integrated, multiscale biophysical and biochemical processes that are currently not well defined. In this brief review, we highlight recent computational and experimental models of cardiac hypertrophy that span mechanisms from the molecular level to the tissue level. Specifically, we focus on: (i) molecular-level models of the structural dynamics of sarcomere proteins in hypertrophic hearts, (ii) cellular-level models of excitation-contraction coupling and mechanosensitive signaling in disease-state myocytes, and (iii) organ-level models of myocardial growth kinematics and predictors thereof. Finally, we discuss how spanning these scales and combining multiple experimental/computational models will provide new information about the processes governing hypertrophy and potential methods to prevent or reverse them.
Collapse
Affiliation(s)
- Yasser Aboelkassem
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Joseph D Powers
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Kimberly J McCabe
- Department of Computational Physiology, Simula Research Laboratory, Lysaker, Norway
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
39
|
Cunningham KS, Spears DA, Care M. Evaluation of cardiac hypertrophy in the setting of sudden cardiac death. Forensic Sci Res 2019; 4:223-240. [PMID: 31489388 PMCID: PMC6713129 DOI: 10.1080/20961790.2019.1633761] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 01/06/2023] Open
Abstract
Ventricular hypertrophy is a common pathological finding at autopsy that can act as a substrate for arrhythmogenesis. Pathologists grapple with the significance of ventricular hypertrophy when assessing the sudden and unexpected deaths of young people and what it could mean for surviving family members. The pathological spectrum of left ventricular hypertrophy (LVH) is reviewed herein. This article is oriented to the practicing autopsy pathologist to help make sense of various patterns of increased heart muscle, particularly those that are not clearly cardiomyopathic, yet present in the setting of sudden cardiac death. The article also reviews factors influencing arrhythmogenesis as well as genetic mutations most commonly associated with ventricular hypertrophy, especially those associated with hypertrophic cardiomyopathy (HCM).
Collapse
Affiliation(s)
- Kristopher S. Cunningham
- Department of Laboratory Medicine and Pathobiology, Ontario Forensic Pathology Service, University of Toronto, Toronto, Canada
| | - Danna A. Spears
- University Health Network, Division of Cardiology – Electrophysiology, University of Toronto, Toronto, Canada
| | - Melanie Care
- Fred A. Litwin Family Centre in Genetic Medicine and Inherited Arrhythmia Clinic, University Health Network & Mount Sinai Hospital, University of Toronto, Toronto, Canada
| |
Collapse
|
40
|
Thin filament dysfunctions caused by mutations in tropomyosin Tpm3.12 and Tpm1.1. J Muscle Res Cell Motil 2019; 41:39-53. [PMID: 31270709 PMCID: PMC7109180 DOI: 10.1007/s10974-019-09532-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022]
Abstract
Tropomyosin is the major regulator of the thin filament. In striated muscle its function is to bind troponin complex and control the access of myosin heads to actin in a Ca2+-dependent manner. It also participates in the maintenance of thin filament length by regulation of tropomodulin and leiomodin, the pointed end-binding proteins. Because the size of the overlap between actin and myosin filaments affects the number of myosin heads which interact with actin, the filament length is one of the determinants of force development. Numerous point mutations in genes encoding tropomyosin lead to single amino acid substitutions along the entire length of the coiled coil that are associated with various types of cardiomyopathy and skeletal muscle disease. Specific regions of tropomyosin interact with different binding partners; therefore, the mutations affect diverse tropomyosin functions. In this review, results of studies on mutations in the genes TPM1 and TPM3, encoding Tpm1.1 and Tpm3.12, are described. The paper is particularly focused on mutation-dependent alterations in the mechanisms of actin-myosin interactions and dynamics of the thin filament at the pointed end.
Collapse
|
41
|
Lehman W, Moore JR, Campbell SG, Rynkiewicz MJ. The Effect of Tropomyosin Mutations on Actin-Tropomyosin Binding: In Search of Lost Time. Biophys J 2019; 116:2275-2284. [PMID: 31130236 PMCID: PMC6588729 DOI: 10.1016/j.bpj.2019.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/06/2019] [Accepted: 05/09/2019] [Indexed: 12/14/2022] Open
Abstract
The initial binding of tropomyosin onto actin filaments and then its polymerization into continuous cables on the filament surface must be precisely tuned to overall thin-filament structure, function, and performance. Low-affinity interaction of tropomyosin with actin has to be sufficiently strong to localize the tropomyosin on actin, yet not so tight that regulatory movement on filaments is curtailed. Likewise, head-to-tail association of tropomyosin molecules must be favorable enough to promote tropomyosin cable formation but not so tenacious that polymerization precedes filament binding. Arguably, little molecular detail on early tropomyosin binding steps has been revealed since Wegner's seminal studies on filament assembly almost 40 years ago. Thus, interpretation of mutation-based actin-tropomyosin binding anomalies leading to cardiomyopathies cannot be described fully. In vitro, tropomyosin binding is masked by explosive tropomyosin polymerization once cable formation is initiated on actin filaments. In contrast, in silico analysis, characterizing molecular dynamics simulations of single wild-type and mutant tropomyosin molecules on F-actin, is not complicated by tropomyosin polymerization at all. In fact, molecular dynamics performed here demonstrates that a midpiece tropomyosin domain is essential for normal actin-tropomyosin interaction and that this interaction is strictly conserved in a number of tropomyosin mutant species. Elsewhere along these mutant molecules, twisting and bending corrupts the tropomyosin superhelices as they "lose their grip" on F-actin. We propose that residual interactions displayed by these mutant tropomyosin structures with actin mimic ones that occur in early stages of thin-filament generation, as if the mutants are recapitulating the assembly process but in reverse. We conclude therefore that an initial binding step in tropomyosin assembly onto actin involves interaction of the essential centrally located domain.
Collapse
Affiliation(s)
- William Lehman
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts.
| | - Jeffrey R Moore
- Department of Biological Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts
| | - Stuart G Campbell
- Departments of Biomedical Engineering and Cellular and Molecular Physiology, Yale University, New Haven, Connecticut
| | - Michael J Rynkiewicz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
42
|
Ishii S, Suzuki M, Ishiwata S, Kawai M. Functional significance of HCM mutants of tropomyosin, V95A and D175N, studied with in vitro motility assays. Biophys Physicobiol 2019; 16:28-40. [PMID: 30923661 PMCID: PMC6435021 DOI: 10.2142/biophysico.16.0_28] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022] Open
Abstract
The majority of hypertrophic cardiomyopathy (HCM) is caused by mutations in sarcomere proteins. We examined tropomyosin (Tpm)’s HCM mutants in humans, V95A and D175N, with in vitro motility assay using optical tweezers to evaluate the effects of the Tpm mutations on the actomyosin interaction at the single molecular level. Thin filaments were reconstituted using these Tpm mutants, and their sliding velocity and force were measured at varying Ca2+ concentrations. Our results indicate that the sliding velocity at pCa ≥8.0 was significantly increased in mutants, which is expected to cause a diastolic problem. The velocity that can be activated by Ca2+ decreased significantly in mutants causing a systolic problem. With sliding force, Ca2+ activatable force decreased in V95A and increased in D175N, which may cause a systolic problem. Our results further demonstrate that the duty ratio determined at the steady state of force generation in saturating [Ca2+] decreased in V95A and increased in D175N. The Ca2+ sensitivity and cooperativity were not significantly affected by the mutations. These results suggest that the two mutants modulate molecular processes of the actomyosin interaction differently, but to result in the same pathology known as HCM.
Collapse
Affiliation(s)
- Shuya Ishii
- Department of Physics, Faculty of Science and Engineering, Waseda University, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Madoka Suzuki
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan.,PRESTO, Japan Science and Technology Agency (JST), Kawaguchi, Saitama 332-0012, Japan
| | - Shin'ichi Ishiwata
- Department of Physics, Faculty of Science and Engineering, Waseda University, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Masataka Kawai
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
43
|
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: 1.3] [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
|
44
|
Sarcomere gene variants act as a genetic trigger underlying the development of left ventricular noncompaction. Pediatr Res 2018; 84:733-742. [PMID: 30188508 DOI: 10.1038/s41390-018-0162-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 06/05/2018] [Accepted: 06/27/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Left ventricular noncompaction (LVNC) is a primary cardiomyopathy with heterogeneous genetic origins. The aim of this study was to elucidate the role of sarcomere gene variants in the pathogenesis and prognosis of LVNC. METHODS AND RESULTS We screened 82 Japanese patients (0-35 years old), with a diagnosis of LVNC, for mutations in seven genes encoding sarcomere proteins, by direct DNA sequencing. We identified variants in a significant proportion of cases (27%), which were associated with poor prognosis (p = 0.012), particularly variants in TPM1, TNNC1, and ACTC1 (p = 0.012). To elucidate the pathological role, we developed and studied human-induced pluripotent stem cells (hiPSCs) from a patient carrying a TPM1 p.Arg178His mutation, who underwent heart transplantation. These cells displayed pathological changes, with mislocalization of tropomyosin 1, causing disruption of the sarcomere structure in cardiomyocytes, and impaired calcium handling. Microarray analysis indicated that the TPM1 mutation resulted in the down-regulation of the expression of numerous genes involved in heart development, and positive regulation of cellular process, especially the calcium signaling pathway. CONCLUSIONS Sarcomere genes are implicated as genetic triggers in the development of LVNC, regulating the expression of numerous genes involved in heart development, or modifying the severity of disease.
Collapse
|
45
|
Matyushenko AM, Koubassova NA, Shchepkin DV, Kopylova GV, Nabiev SR, Nikitina LV, Bershitsky SY, Levitsky DI, Tsaturyan AK. The effects of cardiomyopathy-associated mutations in the head-to-tail overlap junction of α-tropomyosin on its properties and interaction with actin. Int J Biol Macromol 2018; 125:1266-1274. [PMID: 30240712 DOI: 10.1016/j.ijbiomac.2018.09.105] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/15/2018] [Accepted: 09/17/2018] [Indexed: 10/28/2022]
Abstract
Tropomyosin (Tpm) plays a crucial role in the regulation of muscle contraction by controlling actin-myosin interaction. Tpm coiled-coil molecules bind each other via overlap junctions of their N- and C-termini and form a semi-rigid strand that binds the helical surface of an actin filament. The high bending stiffness of the strand is essential for high cooperativity of muscle regulation. Point mutations M8R and K15N in the N-terminal part of the junction and the A277V one in the C-terminal part are associated with dilated cardiomyopathy, while the M281T and I284V mutations are related to hypertrophic cardiomyopathy. To reveal molecular mechanism(s) underlying these pathologies, we studied the properties of recombinant Tpm carrying these mutations using several experimental approaches and molecular dynamic simulation of the junction. The M8R and K15N mutations weakened the interaction between the N- and C-termini of Tpm in the overlap junction and reduced the Tpm affinity for actin. These changes possibly led to a reduction in the regulation cooperativity. The C-terminal mutations caused only small and controversial changes in properties of Tpm and its complex with actin. Their involvement in disease phenotype is possibly caused by interaction with other sarcomere proteins.
Collapse
Affiliation(s)
- Alexander M Matyushenko
- Bach Institute of Biochemistry, Research Centre of Biotechnology of the Russian Academy of Sciences, 33 Leninsky prosp., Moscow 119071, Russia; Institute of Immunology and Physiology, Russian Academy of Sciences, 91 Pervomayskaya ul., Yekaterinburg 620049, Russia
| | - Natalia A Koubassova
- Institute of Immunology and Physiology, Russian Academy of Sciences, 91 Pervomayskaya ul., Yekaterinburg 620049, Russia; Institute of Mechanics, Moscow State University, 1 Mitchurinsky prosp., Moscow 119192, Russia
| | - Daniil V Shchepkin
- Institute of Immunology and Physiology, Russian Academy of Sciences, 91 Pervomayskaya ul., Yekaterinburg 620049, Russia
| | - Galina V Kopylova
- Institute of Immunology and Physiology, Russian Academy of Sciences, 91 Pervomayskaya ul., Yekaterinburg 620049, Russia
| | - Salavat R Nabiev
- Institute of Immunology and Physiology, Russian Academy of Sciences, 91 Pervomayskaya ul., Yekaterinburg 620049, Russia
| | - Larisa V Nikitina
- Institute of Immunology and Physiology, Russian Academy of Sciences, 91 Pervomayskaya ul., Yekaterinburg 620049, Russia
| | - Sergey Y Bershitsky
- Institute of Immunology and Physiology, Russian Academy of Sciences, 91 Pervomayskaya ul., Yekaterinburg 620049, Russia
| | - Dmitrii I Levitsky
- Bach Institute of Biochemistry, Research Centre of Biotechnology of the Russian Academy of Sciences, 33 Leninsky prosp., Moscow 119071, Russia; Belozersky Institute of Physico-Chemical Biology, Moscow State University, 1 Leninskiye Gory bld. 40, Moscow 119234, Russia
| | - Andrey K Tsaturyan
- Institute of Mechanics, Moscow State University, 1 Mitchurinsky prosp., Moscow 119192, Russia.
| |
Collapse
|
46
|
Lehman W, Li X, Kiani FA, Moore JR, Campbell SG, Fischer S, Rynkiewicz MJ. Precise Binding of Tropomyosin on Actin Involves Sequence-Dependent Variance in Coiled-Coil Twisting. Biophys J 2018; 115:1082-1092. [PMID: 30195938 DOI: 10.1016/j.bpj.2018.08.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/08/2018] [Accepted: 08/15/2018] [Indexed: 11/17/2022] Open
Abstract
Often considered an archetypal dimeric coiled coil, tropomyosin nonetheless exhibits distinctive "noncanonical" core residues located at the hydrophobic interface between its component α-helices. Notably, a charged aspartate, D137, takes the place of nonpolar residues otherwise present. Much speculation has been offered to rationalize potential local coiled-coil instability stemming from D137 and its effect on regulatory transitions of tropomyosin over actin filaments. Although experimental approaches such as electron cryomicroscopy reconstruction are optimal for defining average tropomyosin positions on actin filaments, to date, these methods have not captured the dynamics of tropomyosin residues clustered around position 137 or elsewhere. In contrast, computational biochemistry, involving molecular dynamics simulation, is a compelling choice to extend the understanding of local and global tropomyosin behavior on actin filaments at high resolution. Here, we report on molecular dynamics simulation of actin-free and actin-associated tropomyosin, showing noncanonical residue D137 as a locus for tropomyosin twist variation, with marked effects on actin-tropomyosin interactions. We conclude that D137-sponsored coiled-coil twisting is likely to optimize electrostatic side-chain contacts between tropomyosin and actin on the assembled thin filament, while offsetting disparities between tropomyosin pseudorepeat and actin subunit periodicities. We find that D137 has only minor local effects on tropomyosin coiled-coil flexibility, (i.e., on its flexural mobility). Indeed, D137-associated overtwisting may actually augment tropomyosin stiffness on actin filaments. Accordingly, such twisting-induced stiffness of tropomyosin is expected to enhance cooperative regulatory translocation of the tropomyosin cable over actin.
Collapse
Affiliation(s)
- William Lehman
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, Massachusetts.
| | - Xiaochuan Li
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, Massachusetts
| | - Farooq A Kiani
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, Massachusetts
| | - Jeffrey R Moore
- Department of Biological Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts
| | - Stuart G Campbell
- Departments of Biomedical Engineering & Cellular and Molecular Physiology, Yale University, New Haven, Connecticut
| | - Stefan Fischer
- Interdisciplinary Center for Scientific Computing, University of Heidelberg, Heidelberg, Baden-Württemberg, Germany
| | - Michael J Rynkiewicz
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
47
|
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: 1.7] [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
|
48
|
Śliwinska M, Robaszkiewicz K, Czajkowska M, Zheng W, Moraczewska J. Functional effects of substitutions I92T and V95A in actin-binding period 3 of tropomyosin. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1866:558-568. [PMID: 29496559 DOI: 10.1016/j.bbapap.2018.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 02/12/2018] [Accepted: 02/23/2018] [Indexed: 01/10/2023]
Affiliation(s)
- Małgorzata Śliwinska
- Department of Biochemistry and Cell Biology, Faculty of Natural Sciences, Kazimierz Wielki University in Bydgoszcz, Ks. J. Poniatowskiego 12 Str., 85-671 Bydgoszcz, Poland
| | - Katarzyna Robaszkiewicz
- Department of Biochemistry and Cell Biology, Faculty of Natural Sciences, Kazimierz Wielki University in Bydgoszcz, Ks. J. Poniatowskiego 12 Str., 85-671 Bydgoszcz, Poland
| | - Marta Czajkowska
- Department of Biochemistry and Cell Biology, Faculty of Natural Sciences, Kazimierz Wielki University in Bydgoszcz, Ks. J. Poniatowskiego 12 Str., 85-671 Bydgoszcz, Poland
| | - Wenjun Zheng
- Department of Physics, University at Buffalo, SUNY, Buffalo, NY 14260, United States
| | - Joanna Moraczewska
- Department of Biochemistry and Cell Biology, Faculty of Natural Sciences, Kazimierz Wielki University in Bydgoszcz, Ks. J. Poniatowskiego 12 Str., 85-671 Bydgoszcz, Poland.
| |
Collapse
|
49
|
Matyushenko AM, Shchepkin DV, Kopylova GV, Bershitsky SY, Koubassova NA, Tsaturyan AK, Levitsky DI. Functional role of the core gap in the middle part of tropomyosin. FEBS J 2018; 285:871-886. [PMID: 29278453 DOI: 10.1111/febs.14369] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/27/2017] [Accepted: 12/20/2017] [Indexed: 11/30/2022]
Abstract
Tropomyosin (Tpm) is an α-helical coiled-coil actin-binding protein playing an essential role in the regulation of muscle contraction. The middle part of the Tpm molecule has some specific features, such as the presence of noncanonical residues as well as a substantial gap at the interhelical interface, which are believed to destabilize a coiled-coil and impart structural flexibility to this part of the molecule. To study how the gap affects structural and functional properties of α-striated Tpm (the Tpm1.1 isoform that is expressed in cardiac and skeletal muscles) we replaced large conserved apolar core residues located at both sides of the gap with smaller ones by mutations M127A/I130A and M141A/Q144A. We found that in contrast with the stabilizing substitutions D137L and G126R studied earlier, these substitutions have no appreciable influence on thermal unfolding and domain structure of the Tpm molecule. They also do not affect actin-binding properties of Tpm. However, they strongly increase sliding velocity of regulated actin filaments in an in vitro motility assay and cause an oversensitivity of the velocity to Ca2+ similar to the stabilizing substitutions D137L and G126R. Molecular dynamics shows that the substitutions studied here increase bending stiffness of the coiled-coil structure of Tpm, like that of G126R/D137L, probably due to closure of the interhelical gap in the area of the substitutions. Our results clearly indicate that the conserved middle part of Tpm is important for the fine tuning of the Ca2+ regulation of actin-myosin interaction in muscle.
Collapse
Affiliation(s)
- Alexander M Matyushenko
- A. N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Daniil V Shchepkin
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia
| | - Galina V Kopylova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia
| | - Sergey Y Bershitsky
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia
| | | | | | - Dmitrii I Levitsky
- A. N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia.,A. N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Russia
| |
Collapse
|
50
|
Ly T, Krieger I, Tolkatchev D, Krone C, Moural T, Samatey FA, Kang C, Kostyukova AS. Structural destabilization of tropomyosin induced by the cardiomyopathy-linked mutation R21H. Protein Sci 2017; 27:498-508. [PMID: 29105867 DOI: 10.1002/pro.3341] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 11/09/2022]
Abstract
The missense mutation R21H in striated muscle tropomyosin is associated with hypertrophic cardiomyopathy, a genetic cardiac disease and a leading cause of sudden cardiac death in young people. Tropomyosin adopts conformation of a coiled coil which is critical for regulation of muscle contraction. In this study, we investigated the effects of the R21H mutation on the coiled-coil structure of tropomyosin and its interactions with its binding partners, tropomodulin and leiomodin. Using circular dichroism and isothermal titration calorimetry, we found that the mutation profoundly destabilized the structural integrity of αTM1a1-28 Zip, a chimeric peptide containing the first 28 residues of tropomyosin. The mutated αTM1a1-28 Zip was still able to interact with tropomodulin and leiomodin. However, the mutation resulted in a ∼30-fold decrease of αTM1a1-28 Zip's binding affinity to leiomodin. We used a crystal structure of αTM1a1-28 Zip that we solved at 1.5 Å resolution to study the mutation's effect in silico by means of molecular dynamics simulation. The simulation data indicated that while the mutation disrupted αTM1a1-28 Zip's coiled-coil structure, most notably from residue Ala18 to residue His31, it may not affect the N-terminal end of tropomyosin. The drastic decrease of αTM1a1-28 Zip's affinity to leiomodin caused by the mutation may lead to changes in the dynamics at the pointed end of thin filaments. Therefore, the R21H mutation is likely interfering with the regulation of the normal thin filament length essential for proper muscle contraction.
Collapse
Affiliation(s)
- Thu Ly
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington
| | - Inna Krieger
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas
| | - Dmitri Tolkatchev
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington
| | - Cheyenna Krone
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington
| | - Timothy Moural
- Department of Chemistry, Washington State University, Pullman, Washington
| | - Fadel A Samatey
- Trans-Membrane Trafficking Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - ChulHee Kang
- Department of Chemistry, Washington State University, Pullman, Washington
| | - Alla S Kostyukova
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington
| |
Collapse
|