1
|
Gayán-Ordás J, Valverde-Gómez M, Bascompte-Claret R, Sánchez-Flores M, López-Ortega R, Ochoa JP. [Dilated cardiomyopathy and conduction disorder due to TNNI3K mutation]. ARCHIVOS DE CARDIOLOGIA DE MEXICO 2025; 95:253-256. [PMID: 40037387 PMCID: PMC12058104 DOI: 10.24875/acm.24000144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/21/2024] [Indexed: 03/06/2025] Open
Affiliation(s)
- Jara Gayán-Ordás
- Servicio de Cardiología, Hospital Universitario Arnau de Vilanova, Lleida
- Instituto de Recerca Biomèdica de Lleida, Lleida
| | - María Valverde-Gómez
- Servicio de Cardiología, Hospital Universitario 12 de Octubre, Madrid
- Health in Code, A Coruña
| | - Ramón Bascompte-Claret
- Servicio de Cardiología, Hospital Universitario Arnau de Vilanova, Lleida
- Instituto de Recerca Biomèdica de Lleida, Lleida
| | | | - Ricard López-Ortega
- Instituto de Recerca Biomèdica de Lleida, Lleida
- Servicio de Bioquímica Clínica, Hospital Universitario Arnau de Vilanova, Lleida. España
| | | |
Collapse
|
2
|
Micolonghi C, Perrone F, Fabiani M, Caroselli S, Savio C, Pizzuti A, Germani A, Visco V, Petrucci S, Rubattu S, Piane M. Unveiling the Spectrum of Minor Genes in Cardiomyopathies: A Narrative Review. Int J Mol Sci 2024; 25:9787. [PMID: 39337275 PMCID: PMC11431948 DOI: 10.3390/ijms25189787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Hereditary cardiomyopathies (CMPs), including arrhythmogenic cardiomyopathy (ACM), dilated cardiomyopathy (DCM), and hypertrophic cardiomyopathy (HCM), represent a group of heart disorders that significantly contribute to cardiovascular morbidity and mortality and are often driven by genetic factors. Recent advances in next-generation sequencing (NGS) technology have enabled the identification of rare variants in both well-established and minor genes associated with CMPs. Nowadays, a set of core genes is included in diagnostic panels for ACM, DCM, and HCM. On the other hand, despite their lesser-known status, variants in the minor genes may contribute to disease mechanisms and influence prognosis. This review evaluates the current evidence supporting the involvement of the minor genes in CMPs, considering their potential pathogenicity and clinical significance. A comprehensive analysis of databases, such as ClinGen, ClinVar, and GeneReviews, along with recent literature and diagnostic guidelines provides a thorough overview of the genetic landscape of minor genes in CMPs and offers guidance in clinical practice, evaluating each case individually based on the clinical referral, and insights for future research. Given the increasing knowledge on these less understood genetic factors, future studies are essential to clearly assess their roles, ultimately leading to improved diagnostic precision and therapeutic strategies in hereditary CMPs.
Collapse
Affiliation(s)
- Caterina Micolonghi
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Perrone
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Marco Fabiani
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- ALTAMEDICA, Human Genetics, 00198 Rome, Italy
| | - Silvia Caroselli
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Juno Genetics, Reproductive Genetics, 00188 Rome, Italy
| | | | - Antonio Pizzuti
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Aldo Germani
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Vincenzo Visco
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Simona Petrucci
- S. Andrea University Hospital, 00189 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Speranza Rubattu
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Maria Piane
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
3
|
Westbury BC, Watanabe H, Sucov HM. A kinase-dead natural polymorphism in the canine Tnni3k gene. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001164. [PMID: 38828440 PMCID: PMC11140478 DOI: 10.17912/micropub.biology.001164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/15/2024] [Accepted: 05/10/2024] [Indexed: 06/05/2024]
Abstract
Most mammalian cardiomyocytes become polyploid in the neonatal period, concurrent with their loss of proliferative capacity. In mice, natural or engineered mutation of the cardiomyocyte-specific kinase gene Tnni3k causes a higher level of diploid CMs and a higher capacity to support proliferation after adult injury. Here, we identified a polymorphism in the canine Tnni3k gene that is particularly common in the West Highland White Terrier breed, and show that this variant eliminates Tnni3k kinase activity. Thus, in several species, natural Tnni3k polymorphisms exist that are predicted to contribute to variation in diploid CM level and heart regenerative ability.
Collapse
Affiliation(s)
- Baylee C Westbury
- Dept. of Regenerative Medicine and Cell Biology, Medical University of South Carolina
| | - Hirofumi Watanabe
- Dept. of Regenerative Medicine and Cell Biology, Medical University of South Carolina
- Dept. of Pediatrics and Child Health, Nihon Univeristy School of Medicine
| | - Henry M Sucov
- Dept. of Regenerative Medicine and Cell Biology, Medical University of South Carolina
| |
Collapse
|
4
|
Wang L, Sun T, Liu X, Wang Y, Qiao X, Chen N, Liu F, Zhou X, Wang H, Shen H. Myocarditis: A multi-omics approach. Clin Chim Acta 2024; 554:117752. [PMID: 38184138 DOI: 10.1016/j.cca.2023.117752] [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: 12/14/2023] [Revised: 12/29/2023] [Accepted: 12/29/2023] [Indexed: 01/08/2024]
Abstract
Myocarditis, an inflammatory condition of weakened heart muscles often triggered by a variety of causes, that can result in heart failure and sudden death. Novel ways to enhance our understanding of myocarditis pathogenesis is available through newer modalities (omics). In this review, we examine the roles of various biomolecules and associated functional pathways across genomics, transcriptomics, proteomics, and metabolomics in the pathogenesis of myocarditis. Our analysis further explores the reproducibility and variability intrinsic to omics studies, underscoring the necessity and significance of employing a multi-omics approach to gain profound insights into myocarditis pathogenesis. This integrated strategy not only enhances our understanding of the disease, but also confirms the critical importance of a holistic multi-omics approach in disease analysis.
Collapse
Affiliation(s)
- Lulu Wang
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Tao Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Xiaolan Liu
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Yan Wang
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Xiaorong Qiao
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Nuo Chen
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Fangqian Liu
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Xiaoxiang Zhou
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Hua Wang
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Hongxing Shen
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China.
| |
Collapse
|
5
|
Purdy AL, Swift SK, Sucov HM, Patterson M. Tnni3k influences cardiomyocyte S-phase activity and proliferation. J Mol Cell Cardiol 2023; 183:22-26. [PMID: 37597489 PMCID: PMC11645536 DOI: 10.1016/j.yjmcc.2023.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/25/2023] [Accepted: 08/14/2023] [Indexed: 08/21/2023]
Abstract
Cardiomyocyte proliferation is a difficult phenomenon to capture and prove. Here we employ a retrospective analysis of single cell ventricular suspensions to definitively identify cardiomyocytes that have completed cell division. Through this analysis we determined that the capacity of cardiomyocytes to re-enter the cell cycle and complete cell division after injury are separate and variable traits. Further, we provide evidence that Tnni3k definitively influences both early and final stages of the cell cycle.
Collapse
Affiliation(s)
- Alexandra L Purdy
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Samantha K Swift
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Henry M Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States of America
| | - Michaela Patterson
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America.
| |
Collapse
|
6
|
Liu L, Li RG, Martin JF. An improved method reveals the dual role of Tnni3k in promoting S-phase entry while suppressing cell division in cardiomyocytes. J Mol Cell Cardiol 2023; 183:100-101. [PMID: 37769962 DOI: 10.1016/j.yjmcc.2023.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023]
Affiliation(s)
- Lin Liu
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, 6770 Bertner Avenue, Houston, TX 77030, USA
| | - Rich Gang Li
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, 6770 Bertner Avenue, Houston, TX 77030, USA
| | - James F Martin
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, 6770 Bertner Avenue, Houston, TX 77030, USA; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
7
|
Soonpaa MH, Reuter SP, Castelluccio PF, Field LJ. Musings on intrinsic cardiomyocyte cell cycle activity and myocardial regeneration. J Mol Cell Cardiol 2023; 182:86-91. [PMID: 37517369 PMCID: PMC10530305 DOI: 10.1016/j.yjmcc.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 08/01/2023]
Abstract
Although the myocardial renewal rate in the adult mammalian heart is quite low, recent studies have identified genetic variants which can impact the degree of cardiomyocyte cell cycle reentry. Here we use the compound interest law to model the level of regenerative growth over time in mice exhibiting different rates of cardiomyocyte cell cycle reentry following myocardial injury. The modeling suggests that the limited ability of S-phase adult cardiomyocytes to progress through cytokinesis, rather than the ability to reenter the cell cycle per se, is a major contributor to the low levels of intrinsic regenerative growth in the adult myocardium.
Collapse
Affiliation(s)
- Mark H Soonpaa
- Krannert Cardiovascular Research Center and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, USA
| | - Sean P Reuter
- Krannert Cardiovascular Research Center and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, USA
| | - Peter F Castelluccio
- Krannert Cardiovascular Research Center and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, USA
| | - Loren J Field
- Krannert Cardiovascular Research Center and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, USA.
| |
Collapse
|
8
|
Meade RK, Long JE, Jinich A, Rhee KY, Ashbrook DG, Williams RW, Sassetti CM, Smith CM. Genome-wide screen identifies host loci that modulate Mycobacterium tuberculosis fitness in immunodivergent mice. G3 (BETHESDA, MD.) 2023; 13:jkad147. [PMID: 37405387 PMCID: PMC10468300 DOI: 10.1093/g3journal/jkad147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/05/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023]
Abstract
Genetic differences among mammalian hosts and among strains of Mycobacterium tuberculosis (Mtb) are well-established determinants of tuberculosis (TB) patient outcomes. The advent of recombinant inbred mouse panels and next-generation transposon mutagenesis and sequencing approaches has enabled dissection of complex host-pathogen interactions. To identify host and pathogen genetic determinants of Mtb pathogenesis, we infected members of the highly diverse BXD family of strains with a comprehensive library of Mtb transposon mutants (TnSeq). Members of the BXD family segregate for Mtb-resistant C57BL/6J (B6 or B) and Mtb-susceptible DBA/2J (D2 or D) haplotypes. The survival of each bacterial mutant was quantified within each BXD host, and we identified those bacterial genes that were differentially required for Mtb fitness across BXD genotypes. Mutants that varied in survival among the host family of strains were leveraged as reporters of "endophenotypes," each bacterial fitness profile directly probing specific components of the infection microenvironment. We conducted quantitative trait loci (QTL) mapping of these bacterial fitness endophenotypes and identified 140 host-pathogen QTL (hpQTL). We located a QTL hotspot on chromosome 6 (75.97-88.58 Mb) associated with the genetic requirement of multiple Mtb genes: Rv0127 (mak), Rv0359 (rip2), Rv0955 (perM), and Rv3849 (espR). Together, this screen reinforces the utility of bacterial mutant libraries as precise reporters of the host immunological microenvironment during infection and highlights specific host-pathogen genetic interactions for further investigation. To enable downstream follow-up for both bacterial and mammalian genetic research communities, all bacterial fitness profiles have been deposited into GeneNetwork.org and added into the comprehensive collection of TnSeq libraries in MtbTnDB.
Collapse
Affiliation(s)
- Rachel K Meade
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | - Jarukit E Long
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01655, USA
- Research Animal Diagnostic Services, Charles River Laboratories, Wilmington, MA 01887, USA
| | - Adrian Jinich
- Division of Infectious Diseases, Weill Cornell Medical College, New York, NY 10021, USA
| | - Kyu Y Rhee
- Division of Infectious Diseases, Weill Cornell Medical College, New York, NY 10021, USA
| | - David G Ashbrook
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Christopher M Sassetti
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Clare M Smith
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| |
Collapse
|
9
|
Gu Q, Orgil BO, Bajpai AK, Chen Y, Ashbrook DG, Starlard-Davenport A, Towbin JA, Lebeche D, Purevjav E, Sheng H, Lu L. Expression Levels of the Tnni3k Gene in the Heart Are Highly Associated with Cardiac and Glucose Metabolism-Related Phenotypes and Functional Pathways. Int J Mol Sci 2023; 24:12759. [PMID: 37628941 PMCID: PMC10454158 DOI: 10.3390/ijms241612759] [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: 07/11/2023] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Troponin-I interacting kinase encoded by the TNNI3K gene is expressed in nuclei and Z-discs of cardiomyocytes. Mutations in TNNI3K were identified in patients with cardiac conduction diseases, arrhythmias, and cardiomyopathy. METHODS We performed cardiac gene expression, whole genome sequencing (WGS), and cardiac function analysis in 40 strains of BXD recombinant inbred mice derived from C57BL/6J (B6) and DBA/2J (D2) strains. Expression quantitative trait loci (eQTLs) mapping and gene enrichment analysis was performed, followed by validation of candidate Tnni3k-regulatory genes. RESULTS WGS identified compound splicing and missense T659I Tnni3k variants in the D2 parent and some BXD strains (D allele) and these strains had significantly lower Tnni3k expression than those carrying wild-type Tnni3k (B allele). Expression levels of Tnni3k significantly correlated with multiple cardiac (heart rate, wall thickness, PR duration, and T amplitude) and metabolic (glucose levels and insulin resistance) phenotypes in BXDs. A significant cis-eQTL on chromosome 3 was identified for the regulation of Tnni3k expression. Furthermore, Tnni3k-correlated genes were primarily involved in cardiac and glucose metabolism-related functions and pathways. Genes Nodal, Gnas, Nfkb1, Bmpr2, Bmp7, Smad7, Acvr1b, Acvr2b, Chrd, Tgfb3, Irs1, and Ppp1cb were differentially expressed between the B and D alleles. CONCLUSIONS Compound splicing and T659I Tnni3k variants reduce cardiac Tnni3k expression and Tnni3k levels are associated with cardiac and glucose metabolism-related phenotypes.
Collapse
Affiliation(s)
- Qingqing Gu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, China; (Q.G.); (Y.C.)
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (A.K.B.); (D.G.A.); (A.S.-D.)
| | - Buyan-Ochir Orgil
- The Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (B.-O.O.); (J.A.T.); (E.P.)
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38105, USA
| | - Akhilesh Kumar Bajpai
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (A.K.B.); (D.G.A.); (A.S.-D.)
| | - Yufeng Chen
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, China; (Q.G.); (Y.C.)
| | - David G. Ashbrook
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (A.K.B.); (D.G.A.); (A.S.-D.)
| | - Athena Starlard-Davenport
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (A.K.B.); (D.G.A.); (A.S.-D.)
| | - Jeffrey A. Towbin
- The Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (B.-O.O.); (J.A.T.); (E.P.)
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38105, USA
- Pediatric Cardiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Djamel Lebeche
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Enkhsaikhan Purevjav
- The Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (B.-O.O.); (J.A.T.); (E.P.)
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38105, USA
| | - Hongzhuan Sheng
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, China; (Q.G.); (Y.C.)
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (A.K.B.); (D.G.A.); (A.S.-D.)
| |
Collapse
|
10
|
Pham C, Andrzejczyk K, Jurgens SJ, Lekanne Deprez R, Palm KC, Vermeer AM, Nijman J, Christiaans I, Barge-Schaapveld DQ, van Dessel PF, Beekman L, Choi SH, Lubitz SA, Skoric-Milosavljevic D, van den Bersselaar L, Jansen PR, Copier JS, Ellinor PT, Wilde AA, Bezzina CR, Lodder EM. Genetic Burden of TNNI3K in Diagnostic Testing of Patients With Dilated Cardiomyopathy and Supraventricular Arrhythmias. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:328-336. [PMID: 37199186 PMCID: PMC10426786 DOI: 10.1161/circgen.122.003975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 04/10/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Genetic variants in TNNI3K (troponin-I interacting kinase) have previously been associated with dilated cardiomyopathy (DCM), cardiac conduction disease, and supraventricular tachycardias. However, the link between TNNI3K variants and these cardiac phenotypes shows a lack of consensus concerning phenotype and protein function. METHODS We describe a systematic retrospective study of a cohort of patients undergoing genetic testing for cardiac arrhythmias and cardiomyopathy including TNNI3K. We further performed burden testing of TNNI3K in the UK Biobank. For 2 novel TNNI3K variants, we tested cosegregation. TNNI3K kinase function was estimated by TNNI3K autophosphorylation assays. RESULTS We demonstrate enrichment of rare coding TNNI3K variants in DCM patients in the Amsterdam cohort. In the UK Biobank, we observed an association between TNNI3K missense (but not loss-of-function) variants and DCM and atrial fibrillation. Furthermore, we demonstrate genetic segregation for 2 rare variants, TNNI3K-p.Ile512Thr and TNNI3K-p.His592Tyr, with phenotypes consisting of DCM, cardiac conduction disease, and supraventricular tachycardia, together with increased autophosphorylation. In contrast, TNNI3K-p.Arg556_Asn590del, a likely benign variant, demonstrated depleted autophosphorylation. CONCLUSIONS Our findings demonstrate an increased burden of rare coding TNNI3K variants in cardiac patients with DCM. Furthermore, we present 2 novel likely pathogenic TNNI3K variants with increased autophosphorylation, suggesting that enhanced autophosphorylation is likely to drive pathogenicity.
Collapse
Affiliation(s)
- Caroline Pham
- Department of Experimental Cardiology (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., C.R.B., E.M.L.), Heart Center, Amsterdam UMC location University of Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, the Netherlands (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., A.A.M.W., C.R.B., E.M.L.)
| | - Karolina Andrzejczyk
- Department of Experimental Cardiology (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., C.R.B., E.M.L.), Heart Center, Amsterdam UMC location University of Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, the Netherlands (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., A.A.M.W., C.R.B., E.M.L.)
| | - Sean J. Jurgens
- Department of Experimental Cardiology (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., C.R.B., E.M.L.), Heart Center, Amsterdam UMC location University of Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, the Netherlands (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., A.A.M.W., C.R.B., E.M.L.)
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.J.J., S.A.L., P.T.E.)
- Cardiovascular Research Center, Massachusetts General Hospital, Boston (S.J.J., S.A.L., P.T.E.)
| | - Ronald Lekanne Deprez
- Department of Human Genetics, Amsterdam UMC location University of Amsterdam, the Netherlands (R.L.D., A.M.C.V., J.N., D.S.-M., P.R.J., E.M.L.)
| | - Kaylin C.A. Palm
- Department of Experimental Cardiology (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., C.R.B., E.M.L.), Heart Center, Amsterdam UMC location University of Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, the Netherlands (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., A.A.M.W., C.R.B., E.M.L.)
| | - Alexa M.C. Vermeer
- Department of Human Genetics, Amsterdam UMC location University of Amsterdam, the Netherlands (R.L.D., A.M.C.V., J.N., D.S.-M., P.R.J., E.M.L.)
| | - Janneke Nijman
- Department of Human Genetics, Amsterdam UMC location University of Amsterdam, the Netherlands (R.L.D., A.M.C.V., J.N., D.S.-M., P.R.J., E.M.L.)
| | - Imke Christiaans
- Department of Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (I.C.)
| | | | - Pascal F.H.M. van Dessel
- Department of Cardiology, Thorax Center Twente, Medisch Spectrum Twente (MST), Enschede, the Netherlands (P.F.H.M.v.D.)
| | - Leander Beekman
- Department of Experimental Cardiology (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., C.R.B., E.M.L.), Heart Center, Amsterdam UMC location University of Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, the Netherlands (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., A.A.M.W., C.R.B., E.M.L.)
| | | | - Steven A. Lubitz
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.J.J., S.A.L., P.T.E.)
- Cardiovascular Research Center, Massachusetts General Hospital, Boston (S.J.J., S.A.L., P.T.E.)
| | - Doris Skoric-Milosavljevic
- Department of Human Genetics, Amsterdam UMC location University of Amsterdam, the Netherlands (R.L.D., A.M.C.V., J.N., D.S.-M., P.R.J., E.M.L.)
| | - Lisa van den Bersselaar
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands (L.v.d.B.)
| | - Philip R. Jansen
- Department of Human Genetics, Amsterdam UMC location University of Amsterdam, the Netherlands (R.L.D., A.M.C.V., J.N., D.S.-M., P.R.J., E.M.L.)
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Complex Trait Genetics, the Netherlands (P.R.J.)
| | - Jaël S. Copier
- Department of Experimental Cardiology (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., C.R.B., E.M.L.), Heart Center, Amsterdam UMC location University of Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, the Netherlands (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., A.A.M.W., C.R.B., E.M.L.)
| | - Patrick T. Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.J.J., S.A.L., P.T.E.)
- Cardiovascular Research Center, Massachusetts General Hospital, Boston (S.J.J., S.A.L., P.T.E.)
| | - Arthur A.M. Wilde
- Department of Cardiology (A.A.M.W.), Heart Center, Amsterdam UMC location University of Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, the Netherlands (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., A.A.M.W., C.R.B., E.M.L.)
| | - Connie R. Bezzina
- Department of Experimental Cardiology (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., C.R.B., E.M.L.), Heart Center, Amsterdam UMC location University of Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, the Netherlands (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., A.A.M.W., C.R.B., E.M.L.)
| | - Elisabeth M. Lodder
- Department of Experimental Cardiology (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., C.R.B., E.M.L.), Heart Center, Amsterdam UMC location University of Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, the Netherlands (C.P., K.A., S.J.J., K.C.A.P., L.B., J.S.C., A.A.M.W., C.R.B., E.M.L.)
- Department of Human Genetics, Amsterdam UMC location University of Amsterdam, the Netherlands (R.L.D., A.M.C.V., J.N., D.S.-M., P.R.J., E.M.L.)
| |
Collapse
|
11
|
Ding Y, Wang M, Bu H, Li J, Lin X, Xu X. Application of an F0-based genetic assay in adult zebrafish to identify modifier genes of an inherited cardiomyopathy. Dis Model Mech 2023; 16:dmm049427. [PMID: 35481478 PMCID: PMC9239171 DOI: 10.1242/dmm.049427] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/19/2022] [Indexed: 01/08/2023] Open
Abstract
Modifier genes contribute significantly to our understanding of pathophysiology in human diseases; however, effective approaches to identify modifier genes are still lacking. Here, we aim to develop a rapid F0-based genetic assay in adult zebrafish using the bag3 gene knockout (bag3e2/e2) cardiomyopathy model as a paradigm. First, by utilizing a classic genetic breeding approach, we identified dnajb6b as a deleterious modifier gene for bag3 cardiomyopathy. Next, we established an F0-based genetic assay in adult zebrafish through injection of predicted microhomology-mediated end joining (MMEJ)-inducing single guide RNA/Cas9 protein complex. We showed that effective gene knockdown is maintained in F0 adult fish, enabling recapitulation of both salutary modifying effects of the mtor haploinsufficiency and deleterious modifying effects of the dnajb6b gene on bag3 cardiomyopathy. We finally deployed the F0-based genetic assay to screen differentially expressed genes in the bag3 cardiomyopathy model. As a result, myh9b was identified as a novel modifier gene for bag3 cardiomyopathy. Together, these data prove the feasibility of an F0 adult zebrafish-based genetic assay that can be effectively used to discover modifier genes for inherited cardiomyopathy.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Mingmin Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Haisong Bu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiothoracic Surgery, Xiangfan Hospital, Central South University, Changsha 410008, China
| | - Jiarong Li
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Surgery, The Second Xiangfan Hospital of Central South University, Changsha 410011, China
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
12
|
Swift SK, Purdy AL, Kolell ME, Andresen KG, Lahue C, Buddell T, Akins KA, Rau CD, O'Meara CC, Patterson M. Cardiomyocyte ploidy is dynamic during postnatal development and varies across genetic backgrounds. Development 2023; 150:dev201318. [PMID: 36912240 PMCID: PMC10113957 DOI: 10.1242/dev.201318] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 03/06/2023] [Indexed: 03/14/2023]
Abstract
Somatic polyploidization, an adaptation by which cells increase their DNA content to support growth, is observed in many cell types, including cardiomyocytes. Although polyploidization is believed to be beneficial, progression to a polyploid state is often accompanied by loss of proliferative capacity. Recent work suggests that genetics heavily influence cardiomyocyte ploidy. However, the developmental course by which cardiomyocytes reach their final ploidy state has only been investigated in select backgrounds. Here, we assessed cardiomyocyte number, cell cycle activity, and ploidy dynamics across two divergent mouse strains: C57BL/6J and A/J. Both strains are born and reach adulthood with comparable numbers of cardiomyocytes; however, the end composition of ploidy classes and developmental progression to reach the final state differ substantially. We expand on previous findings that identified Tnni3k as a mediator of cardiomyocyte ploidy and uncover a role for Runx1 in ploidy dynamics and cardiomyocyte cell division, in both developmental and injury contexts. These data provide novel insights into the developmental path to cardiomyocyte polyploidization and challenge the paradigm that hypertrophy is the sole mechanism for growth in the postnatal heart.
Collapse
Affiliation(s)
- Samantha K Swift
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
| | - Alexandra L Purdy
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
| | - Mary E Kolell
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
| | - Kaitlyn G Andresen
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
| | - Caitlin Lahue
- University of North Carolina School of Medicine, Department of Genetics, Chapel Hill, NC 27599, USA
| | - Tyler Buddell
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
- Medical College of Wisconsin, Cardiovascular Center, Milwaukee, WI 53226, USA
| | - Kaelin A Akins
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
| | - Christoph D Rau
- University of North Carolina School of Medicine, Department of Genetics, Chapel Hill, NC 27599, USA
| | - Caitlin C O'Meara
- Medical College of Wisconsin, Cardiovascular Center, Milwaukee, WI 53226, USA
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI 53226, USA
| | - Michaela Patterson
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
- Medical College of Wisconsin, Cardiovascular Center, Milwaukee, WI 53226, USA
| |
Collapse
|
13
|
Embryonic Hyperglycemia Disrupts Myocardial Growth, Morphological Development, and Cellular Organization: An In Vivo Experimental Study. Life (Basel) 2023; 13:life13030768. [PMID: 36983924 PMCID: PMC10056749 DOI: 10.3390/life13030768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023] Open
Abstract
Hyperglycemia during gestation can disrupt fetal heart development and increase postnatal cardiovascular disease risk. It is therefore imperative to identify early biomarkers of hyperglycemia during gestation-induced fetal heart damage and elucidate the underlying molecular pathomechanisms. Clinical investigations of diabetic adults with heart dysfunction and transgenic mouse studies have revealed that overexpression or increased expression of TNNI3K, a heart-specific kinase that binds troponin cardiac I, may contribute to abnormal cardiac remodeling, ventricular hypertrophy, and heart failure. Optimal heart function also depends on the precise organization of contractile and excitable tissues conferred by intercellular occlusive, adherent, and communicating junctions. The current study evaluated changes in embryonic heart development and the expression levels of sarcomeric proteins (troponin I, desmin, and TNNI3K), junctional proteins, glucose transporter-1, and Ki-67 under fetal hyperglycemia. Stage 22HH Gallus domesticus embryos were randomly divided into two groups: a hyperglycemia (HG) group, in which individual embryos were injected with 30 mmol/L glucose solution every 24 h for 10 days, and a no-treatment (NT) control group, in which individual embryos were injected with physiological saline every 24 h for 10 days (stage 36HH). Embryonic blood glucose, height, and weight, as well as heart size, were measured periodically during treatment, followed by histopathological analysis and estimation of sarcomeric and junctional protein expression by western blotting and immunostaining. Hyperglycemic embryos demonstrated delayed heart maturation, with histopathological analysis revealing reduced left and right ventricular wall thickness (−39% and −35% vs. NT). Immunoexpression levels of TNNI3K and troponin 1 increased (by 37% and 39%, respectively), and desmin immunofluorescence reduced (by 23%). Embryo-fetal hyperglycemia may trigger an increase in the expression levels of TNNI3K and troponin I, as well as dysfunction of occlusive and adherent junctions, ultimately inducing abnormal cardiac remodeling.
Collapse
|
14
|
Ding D, Braun T. A Tedious Journey: Cardiomyocyte Proliferation Requires More Than S-Phase Entry and Loss of Polyploidization. Circulation 2023; 147:154-157. [PMID: 36622907 DOI: 10.1161/circulationaha.122.062784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Dong Ding
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (D.D., T.B.)
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (D.D., T.B.).,German Center for Cardiovascular Research (DZHK), Rhine Main, Germany (T.B.).,German Center for Lung Research (DZL), Giessen, Germany (T.B.)
| |
Collapse
|
15
|
Reuter SP, Soonpaa MH, Field D, Simpson E, Rubart-von der Lohe M, Lee HK, Sridhar A, Ware SM, Green N, Li X, Ofner S, Marchuk DA, Wollert KC, Field LJ. Cardiac Troponin I-Interacting Kinase Affects Cardiomyocyte S-Phase Activity but Not Cardiomyocyte Proliferation. Circulation 2023; 147:142-153. [PMID: 36382596 PMCID: PMC9839600 DOI: 10.1161/circulationaha.122.061130] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Identifying genetic variants that affect the level of cell cycle reentry and establishing the degree of cell cycle progression in those variants could help guide development of therapeutic interventions aimed at effecting cardiac regeneration. We observed that C57Bl6/NCR (B6N) mice have a marked increase in cardiomyocyte S-phase activity after permanent coronary artery ligation compared with infarcted DBA/2J (D2J) mice. METHODS Cardiomyocyte cell cycle activity after infarction was monitored in D2J, (D2J×B6N)-F1, and (D2J×B6N)-F1×D2J backcross mice by means of bromodeoxyuridine or 5-ethynyl-2'-deoxyuridine incorporation using a nuclear-localized transgenic reporter to identify cardiomyocyte nuclei. Genome-wide quantitative trait locus analysis, fine scale genetic mapping, whole exome sequencing, and RNA sequencing analyses of the backcross mice were performed to identify the gene responsible for the elevated cardiomyocyte S-phase phenotype. RESULTS (D2J×B6N)-F1 mice exhibited a 14-fold increase in cardiomyocyte S-phase activity in ventricular regions remote from infarct scar compared with D2J mice (0.798±0.09% versus 0.056±0.004%; P<0.001). Quantitative trait locus analysis of (D2J×B6N)-F1×D2J backcross mice revealed that the gene responsible for differential S-phase activity was located on the distal arm of chromosome 3 (logarithm of the odds score=6.38; P<0.001). Additional genetic and molecular analyses identified 3 potential candidates. Of these, Tnni3k (troponin I-interacting kinase) is expressed in B6N hearts but not in D2J hearts. Transgenic expression of TNNI3K in a D2J genetic background results in elevated cardiomyocyte S-phase activity after injury. Cardiomyocyte S-phase activity in both Tnni3k-expressing and Tnni3k-nonexpressing mice results in the formation of polyploid nuclei. CONCLUSIONS These data indicate that Tnni3k expression increases the level of cardiomyocyte S-phase activity after injury.
Collapse
Affiliation(s)
- Sean P. Reuter
- Krannert Cardiovascular Research Center, Indiana University School of Medicine
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Mark H. Soonpaa
- Krannert Cardiovascular Research Center, Indiana University School of Medicine
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Dorothy Field
- Krannert Cardiovascular Research Center, Indiana University School of Medicine
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Ed Simpson
- Center for Computational Biology & Bioinformatics, Indiana University School of Medicine
| | | | - Han Kyu Lee
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine
| | - Arthi Sridhar
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Stephanie M. Ware
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Nick Green
- Center for Computational Biology & Bioinformatics, Indiana University School of Medicine
| | - Xiaochun Li
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine
| | - Susan Ofner
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine
| | - Douglas A. Marchuk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine
| | - Kai C. Wollert
- Department of Cardiology and Angiology, Division of Molecular and Translational Cardiology, Hannover Medical School
| | - Loren J. Field
- Krannert Cardiovascular Research Center, Indiana University School of Medicine
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| |
Collapse
|
16
|
Qu H, Zhang Y, Zhang W, Zhu Y, Xu R. Knockout of Cardiac Troponin I-Interacting Kinase leads to cardiac dysfunction and remodeling. Clin Exp Pharmacol Physiol 2022; 49:1169-1178. [PMID: 35781726 DOI: 10.1111/1440-1681.13699] [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: 02/02/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022]
Abstract
Cardiac troponin I-interacting kinase (TNNI3K) is a cardiac-specific kinase that has been identified as a diagnostic marker and a therapeutic target in cardiovascular diseases. However, the biological function of TNNI3K in cardiac dysfunction and remodeling remain elusive. In the present study, a Tnni3k cardiomyocyte-specific knockout (Tnni3k-cKO) mouse model was established. Echocardiography was used to evaluate cardiac function in mice. Heart failure markers were detected using enzyme-linked immunosorbent assay. Hematoxylin and eosin staining, wheat germ agglutinin staining, Masson's trichrome staining, Sirius red staining, and TUNEL staining were used to assess histopathological changes, cardiac hypertrophy, collagen deposition, and myocardial apoptosis, respectively. Expression levels of TNNI3K, apoptosis-related proteins, and p38 mitogen-activated protein kinase were measured using Western blot analysis. Compared to wild-type controls, cardiac dysfunction and cardiac remodeling of Tnni3k-cKO mice increased gradually with age. Tnni3k-cKO mice exhibited cardiac hypertrophy, cardiac fibrosis, and cardiomyocyte apoptosis. Upregulation of cleaved caspase-3 in Tnni3k-cKO mice appeared to be related to phosphorylation and activation of the p38 mitogen-activated protein kinase signaling pathway. In conclusion, this study shows that TNNI3K is essential for cardiac development and function, providing new insights into the development of novel therapeutic strategies for cardiac diseases.
Collapse
Affiliation(s)
- Huilin Qu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Zhang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Zhang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yaxin Zhu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruixia Xu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
Xie T, Yang Y, Gong K, Luo Y, Guo H, Liu R, Wang L, Tan Z, Luo J, Xie L. Whole-Exome Sequencing Identifies a Novel Variant (c.1538T > C) of TNNI3K in Arrhythmogenic Right Ventricular Cardiomyopathy. Front Cardiovasc Med 2022; 9:843837. [PMID: 35274013 PMCID: PMC8902045 DOI: 10.3389/fcvm.2022.843837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/25/2022] [Indexed: 11/25/2022] Open
Abstract
Backgrounds Arrhythmic right ventricular cardiomyopathy (ARVC) is a cardiomyopathy with a genetic predisposition that can lead to a sudden cardiac death and heart failure. According to the 2010 Task Force Criteria, genetic diagnosis is one of the most important methods, but, so far, only a few genes related to ARVC have been identified. Methods In this study, the pathogenic gene of a patient with ARVC was examined using whole-exome sequencing. The plasmids of TNNI3K were constructed, and the effects of the TNNI3K variant was investigated by a real-time polymerase chain reaction (PCR) and western blot. Results A novel variant (c.1538T > C) of TNNI3K was identified, with phenotypes of dominant right ventricular (RV) disease preliminarily fulfilling the diagnosis of ARVC. A comprehensive assessment revealed that the variant was pathogenic. We found that this variant would lead to a decrease in the level of TNNI3K mRNA and protein, as well as a decrease in the expression of the RYR2 gene, which further proves that TNNI3K plays an important role in cardiomyopathy and expands the spectrum of the TNNI3K variants. Conclusion In this study, we reported a TNNI3K variant in ARVC for the first time, and the results not only contribute to the diagnosis of ARVC, but also provide a reference for genetic counseling and promote the understanding of the genetic mechanism of cardiomyopathy.
Collapse
Affiliation(s)
- Ting Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
- The Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
- The Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Ke Gong
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
- The Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Yong Luo
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
- The Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Hui Guo
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
- The Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Ruilin Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
- The Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Lei Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
- The Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Zhiping Tan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
- The Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Jinwen Luo
- Department of Cardio-Thoracic Surgery, Hunan Children's Hospital, Changsha, China
| | - Li Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
- The Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
- *Correspondence: Li Xie
| |
Collapse
|
18
|
Patterson JR, Graves AP, Stoy P, Cheung M, Desai TA, Fries H, Gatto GJ, Holt DA, Shewchuk L, Totoritis R, Wang L, Kallander LS. Identification of Diarylurea Inhibitors of the Cardiac-Specific Kinase TNNI3K by Designing Selectivity Against VEGFR2, p38α, and B-Raf. J Med Chem 2021; 64:15651-15670. [PMID: 34699203 DOI: 10.1021/acs.jmedchem.1c00700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A series of diarylurea inhibitors of the cardiac-specific kinase TNNI3K were developed to elucidate the biological function of TNNI3K and evaluate TNNI3K as a therapeutic target for the treatment of cardiovascular diseases. Utilizing a structure-based design, enhancements in kinase selectivity were engineered into the series, capitalizing on the established X-ray crystal structures of TNNI3K, VEGFR2, p38α, and B-Raf. Our efforts culminated in the discovery of an in vivo tool compound 47 (GSK329), which exhibited desirable TNNI3K potency and rat pharmacokinetic properties as well as promising kinase selectivity against VEGFR2 (40-fold), p38α (80-fold), and B-Raf (>200-fold). Compound 47 demonstrated positive cardioprotective outcomes in a mouse model of ischemia/reperfusion cardiac injury, indicating that optimized exemplars from this series, such as 47, are favorable leads for discovering novel medicines for cardiac diseases.
Collapse
Affiliation(s)
- Jaclyn R Patterson
- Heart Failure Discovery Performance Unit, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Alan P Graves
- Platform Technology and Sciences, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Patrick Stoy
- Heart Failure Discovery Performance Unit, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Mui Cheung
- Heart Failure Discovery Performance Unit, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Tina A Desai
- Heart Failure Discovery Performance Unit, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Harvey Fries
- Heart Failure Discovery Performance Unit, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Gregory J Gatto
- Heart Failure Discovery Performance Unit, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Dennis A Holt
- Heart Failure Discovery Performance Unit, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Lisa Shewchuk
- Platform Technology and Sciences, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Rachel Totoritis
- Platform Technology and Sciences, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Liping Wang
- Platform Technology and Sciences, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Lara S Kallander
- Heart Failure Discovery Performance Unit, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| |
Collapse
|
19
|
Pham C, Muñoz-Martín N, Lodder EM. The Diverse Roles of TNNI3K in Cardiac Disease and Potential for Treatment. Int J Mol Sci 2021; 22:6422. [PMID: 34203974 PMCID: PMC8232738 DOI: 10.3390/ijms22126422] [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: 05/25/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/25/2022] Open
Abstract
In the two decades since the discovery of TNNI3K it has been implicated in multiple cardiac phenotypes and physiological processes. TNNI3K is an understudied kinase, which is mainly expressed in the heart. Human genetic variants in TNNI3K are associated with supraventricular arrhythmias, conduction disease, and cardiomyopathy. Furthermore, studies in mice implicate the gene in cardiac hypertrophy, cardiac regeneration, and recovery after ischemia/reperfusion injury. Several new papers on TNNI3K have been published since the last overview, broadening the clinical perspective of TNNI3K variants and our understanding of the underlying molecular biology. We here provide an overview of the role of TNNI3K in cardiomyopathy and arrhythmia covering both a clinical perspective and basic science advancements. In addition, we review the potential of TNNI3K as a target for clinical treatments in different cardiac diseases.
Collapse
Affiliation(s)
| | | | - Elisabeth M. Lodder
- Department of Clinical and Experimental Cardiology, Heart Center, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (C.P.); (N.M.-M.)
| |
Collapse
|
20
|
Touat-Hamici Z, Blancard M, Ma R, Lin L, Iddir Y, Denjoy I, Leenhardt A, Yuchi Z, Guicheney P. A SPRY1 domain cardiac ryanodine receptor variant associated with short-coupled torsade de pointes. Sci Rep 2021; 11:5243. [PMID: 33664309 PMCID: PMC7970841 DOI: 10.1038/s41598-021-84373-9] [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: 06/24/2020] [Accepted: 02/03/2021] [Indexed: 12/15/2022] Open
Abstract
Idiopathic ventricular fibrillation (IVF) causes sudden death in young adult patients without structural or ischemic heart disease. Most IVF cases are sporadic and some patients present with short-coupled torsade de pointes, the genetics of which are poorly understood. A man who had a first syncope at the age of 35 presented with frequent short-coupled premature ventricular beats with bursts of polymorphic ventricular tachycardia and then died suddenly. By exome sequencing, we identified three rare variants: p.I784F in the SPRY1 of the ryanodine receptor 2 (RyR2), p.A96S in connexin 40 (Cx40), reported to affect electrical coupling and cardiac conduction, and a nonsense p.R244X in the cardiac-specific troponin I-interacting kinase (TNNI3K). We assessed intracellular Ca2+ handling in WT and mutant human RYR2 transfected HEK293 cells by fluorescent microscopy and an enhanced store overload-induced Ca2+ release in response to cytosolic Ca2+ was observed in RyR2-I784F cells. In addition, crystal structures and thermal melting temperatures revealed a conformational change in the I784F-SPRY1 domain compared to the WT-domain. The novel RyR2-I784F variant in SPRY1 domain causes a leaky channel under non-stress conditions. The presence of several variants affecting Ca2+ handling and cardiac conduction suggests a possible oligogenic origin for the ectopies originating from Purkinje fibres.
Collapse
Affiliation(s)
- Zahia Touat-Hamici
- INSERM, UMRS 1166, Faculté de Médecine Sorbonne-Université, Unité de Recherche sur les Maladies Cardiovasculaires et Métaboliques, 91, boulevard de l'Hôpital, 75013, Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Malorie Blancard
- INSERM, UMRS 1166, Faculté de Médecine Sorbonne-Université, Unité de Recherche sur les Maladies Cardiovasculaires et Métaboliques, 91, boulevard de l'Hôpital, 75013, Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Paris, France
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ruifang Ma
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Lianyun Lin
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Yasmine Iddir
- INSERM, UMRS 1166, Faculté de Médecine Sorbonne-Université, Unité de Recherche sur les Maladies Cardiovasculaires et Métaboliques, 91, boulevard de l'Hôpital, 75013, Paris, France
- Département d'Oncologie Pédiatrique Laboratoire RTOP «Recherche Translationnelle en Oncologie Pédiatrique»-INSERM U830, Institut Curie, Paris, France
| | - Isabelle Denjoy
- Département de Cardiologie et Centre de Référence des Maladies Cardiaques Héréditaires, AP-HP, Hôpital Bichat, 75018, Paris, France
- Université de Paris, INSERM, U1166, 75013, Paris, France
| | - Antoine Leenhardt
- Département de Cardiologie et Centre de Référence des Maladies Cardiaques Héréditaires, AP-HP, Hôpital Bichat, 75018, Paris, France
- Université de Paris, INSERM, U1166, 75013, Paris, France
| | - Zhiguang Yuchi
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China.
| | - Pascale Guicheney
- INSERM, UMRS 1166, Faculté de Médecine Sorbonne-Université, Unité de Recherche sur les Maladies Cardiovasculaires et Métaboliques, 91, boulevard de l'Hôpital, 75013, Paris, France.
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Paris, France.
| |
Collapse
|
21
|
Gan P, Baicu C, Watanabe H, Wang K, Tao G, Judge DP, Zile MR, Makita T, Mukherjee R, Sucov HM. The prevalent I686T human variant and loss-of-function mutations in the cardiomyocyte-specific kinase gene TNNI3K cause adverse contractility and concentric remodeling in mice. Hum Mol Genet 2020; 29:3504-3515. [PMID: 33084860 DOI: 10.1093/hmg/ddaa234] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/18/2020] [Accepted: 10/14/2020] [Indexed: 01/04/2023] Open
Abstract
TNNI3K expression worsens disease progression in several mouse heart pathology models. TNNI3K expression also reduces the number of diploid cardiomyocytes, which may be detrimental to adult heart regeneration. However, the gene is evolutionarily conserved, suggesting a beneficial function that has remained obscure. Here, we show that C57BL/6J-inbred Tnni3k mutant mice develop concentric remodeling, characterized by ventricular wall thickening and substantial reduction of cardiomyocyte aspect ratio. This pathology occurs in mice carrying a Tnni3k null allele, a K489R point mutation rendering the protein kinase-dead, or an allele corresponding to human I686T, the most common human non-synonymous TNNI3K variant, which is hypomorphic for kinase activity. Mutant mice develop these conditions in the absence of fibrosis or hypertension, implying a primary cardiomyocyte etiology. In culture, mutant cardiomyocytes were impaired in contractility and calcium dynamics and in protein kinase A signaling in response to isoproterenol, indicating diminished contractile reserve. These results demonstrate a beneficial function of TNNI3K in the adult heart that might explain its evolutionary conservation and imply that human TNNI3K variants, in particular the widespread I686T allele, may convey elevated risk for altered heart geometry and hypertrophy.
Collapse
Affiliation(s)
- Peiheng Gan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA.,Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Catalin Baicu
- Department of Medicine Division of Cardiology, Medical University of South Carolina, Charleston, SC, USA
| | - Hirofumi Watanabe
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Kristy Wang
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Daniel P Judge
- Department of Medicine Division of Cardiology, Medical University of South Carolina, Charleston, SC, USA
| | - Michael R Zile
- Department of Medicine Division of Cardiology, Medical University of South Carolina, Charleston, SC, USA
| | - Takako Makita
- Darby Children's Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Rupak Mukherjee
- Department of Medicine Division of Cardiology, Medical University of South Carolina, Charleston, SC, USA
| | - Henry M Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA.,Department of Medicine Division of Cardiology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
22
|
Liu J, Liu D, Li M, Wu K, Liu N, Zhao C, Shi X, Liu Q. Identification of a nonsense mutation in TNNI3K associated with cardiac conduction disease. J Clin Lab Anal 2020; 34:e23418. [PMID: 32529721 PMCID: PMC7521241 DOI: 10.1002/jcla.23418] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cardiac conduction disease (CCD) is a common cardiovascular disease which can lead to life-threatening conditions. The importance of heredity in CCD has been realized in recent years. Several causal genes have been found to be implicated in CCD such as SCN5A, TRPM4, SCN1B, TNNI3K, LMNA, and NKX2.5. To date, only four genetic mutations in TNNI3K have been identified related to CCD. METHODS Whole-exome sequencing (WES) was carried out in order to identify the underlying disease-causing mutation in a Chinese family with CCD. The potential mutations were confirmed by Sanger sequencing. Real-time qPCR was used to detect the level of TNNI3K mRNA expression. RESULTS A nonsense mutation in TNNI3K (NM_015978.2: g.170891C > T, c.1441C > T) was identified in this family and validated by Sanger sequencing. Real-time qPCR confirmed that the level of TNNI3K mRNA expression was decreased compared with the controls. CONCLUSIONS This study found the first nonsense TNNI3K mutation associated with CCD in a Chinese family. TNNI3K harboring the mutation (c.1441C > T) implicated a loss-of-function pathogenic mechanism with an autosomal dominant inheritance pattern. This research enriches the phenotypic spectrum of TNNI3K mutations, casting a new light upon the genotype-phenotype correlations between TNNI3K mutations and CCD and indicating the importance of TNNI3K screening in CCD patients.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Da Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Muzheng Li
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Keke Wu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Na Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chenyu Zhao
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoliu Shi
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China.,Department of Medical Genetics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qiming Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
23
|
New Insights into 4-Anilinoquinazolines as Inhibitors of Cardiac Troponin I-Interacting Kinase (TNNi3K). Molecules 2020; 25:molecules25071697. [PMID: 32272798 PMCID: PMC7180948 DOI: 10.3390/molecules25071697] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 11/16/2022] Open
Abstract
We report the synthesis of several related 4-anilinoquinazolines as inhibitors of cardiac troponin I-interacting kinase (TNNi3K). These close structural analogs of 3-((6,7-dimethoxyquinazolin-4-yl)amino)-4-(dimethylamino)-N-methylbenzenesulfonamide (GSK114) provide new understanding of structure-activity relationships between the 4-anilinoquinazoline scaffold and TNNi3K inhibition. Through a small focused library of inhibitors, we observed that the N-methylbenzenesulfonamide was driving the potency in addition to the more traditional quinazoline hinge-binding motif. We also identified a compound devoid of TNNi3K kinase activity due to the addition of a methyl group in the hinge binding region. This compound could serve as a negative control in the study of TNNi3K biology. Small molecule crystal structures of several quinazolines have been solved, supporting observations made about overall conformation and TNNi3K inhibition.
Collapse
|
24
|
Balasubramanian PK, Balupuri A, Bhujbal SP, Cho SJ. 3D-QSAR Assisted Design of Novel 7-Deazapurine Derivatives as TNNI3K Kinase Inhibitors Using Molecular Docking and Molecular Dynamics Simulation. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180816666190110121300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Cardiac troponin I-interacting kinase (TNNI3K) is a cardiac-specific kinase
that belongs to MAPKKK family. It is a dual-function kinase with tyrosine and serine/threonine
kinase activity. Over-expression of TNNI3K results in various cardiovascular diseases such as
cardiomyopathy, ischemia/reperfusion injury, heart failure, etc. Since, it is a cardiac-specific kinase
and expressed only in heart tissue, it is an ideal molecular target to treat cardiac diseases. The main
objective of the work is to study and understand the structure-activity relationship of the reported
deazapurine derivatives and to use the 3D-QSAR and docking results to design potent and novel
TNNI3K inhibitors of this series.
Methods:
In the present study, we have used molecular docking 3D QSAR, and molecular dynamics
simulation to understand the structure-activity correlation of reported TNNI3K inhibitors and to
design novel compounds of deazapurine derivatives with increased activity.
Results:
Both CoMFA (q2=0.669, NOC=5, r2=0.944) and CoMSIA (q2=0.783, NOC=5, r2=0.965)
have resulted in satisfactory models. The models were validated using external test set, Leave-out-
Five, bootstrapping, progressive scrambling, and rm2 metrics calculations. The validation procedures
showed the developed models were robust and reliable. The docking results and the contour maps
analysis helped in the better understanding of the structure-activity relationship.
Conclusion:
This is the first report on 3D-QSAR modeling studies of TNNI3K inhibitors. Both
docking and MD results were consistent and showed good correlation with the previous experimental
data. Based on the information obtained from contour maps, 31 novel TNNI3K inhibitors were
designed. These designed compounds showed higher activity than the existing dataset compounds.
Collapse
Affiliation(s)
| | - Anand Balupuri
- Department of Biomedical Sciences, College of Medicine, Chosun University, Gwangju 501-759, Korea
| | - Swapnil P. Bhujbal
- Department of Biomedical Sciences, College of Medicine, Chosun University, Gwangju 501-759, Korea
| | - Seung Joo Cho
- Department of Biomedical Sciences, College of Medicine, Chosun University, Gwangju 501-759, Korea
| |
Collapse
|
25
|
Tnni3k alleles influence ventricular mononuclear diploid cardiomyocyte frequency. PLoS Genet 2019; 15:e1008354. [PMID: 31589606 PMCID: PMC6797218 DOI: 10.1371/journal.pgen.1008354] [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: 06/10/2019] [Revised: 10/17/2019] [Accepted: 08/07/2019] [Indexed: 12/29/2022] Open
Abstract
Recent evidence implicates mononuclear diploid cardiomyocytes as a proliferative and regenerative subpopulation of the postnatal heart. The number of these cardiomyocytes is a complex trait showing substantial natural variation among inbred mouse strains based on the combined influences of multiple polymorphic genes. One gene confirmed to influence this parameter is the cardiomyocyte-specific kinase Tnni3k. Here, we have studied Tnni3k alleles across a number of species. Using a newly-generated kinase-dead allele in mice, we show that Tnni3k function is dependent on its kinase activity. In an in vitro kinase assay, we show that several common human TNNI3K kinase domain variants substantially compromise kinase activity, suggesting that TNNI3K may influence human heart regenerative capacity and potentially also other aspects of human heart disease. We show that two kinase domain frameshift mutations in mice cause loss-of-function consequences by nonsense-mediated decay. We further show that the Tnni3k gene in two species of mole-rat has independently devolved into a pseudogene, presumably associated with the transition of these species to a low metabolism and hypoxic subterranean life. This may be explained by the observation that Tnni3k function in mice converges with oxidative stress to regulate mononuclear diploid cardiomyocyte frequency. Unlike other studied rodents, naked mole-rats have a surprisingly high (30%) mononuclear cardiomyocyte level but most of their mononuclear cardiomyocytes are polyploid; their mononuclear diploid cardiomyocyte level (7%) is within the known range (2–10%) of inbred mouse strains. Naked mole-rats provide further insight on a recent proposal that cardiomyocyte polyploidy is associated with evolutionary acquisition of endothermy. Embryonic cardiomyocytes have one diploid nucleus (like most cells of the body), but most adult cardiomyocytes are polyploid. Most adult cardiomyocytes are also post-mitotic and nonregenerative, and as a result, heart injury (such as from a heart attack) is followed by scarring and impaired function rather than by regeneration. A subset of cardiomyocytes in the adult heart remains mononuclear diploid, and recent evidence indicates that this subpopulation has proliferative and regenerative capacity. Our previous work in mice showed that the percentage of this cell population in the adult heart is a complex trait subject to the combined influence of a number of polymorphic genes. One gene that influences variation in this trait is a kinase gene known as Tnni3k. This study addresses the consequences of a number of Tnni3k alleles, both newly engineered in mice and naturally occurring in a number of species, including human and mole-rat, and studied at the phenotypic and biochemical level. These results provide insight into inter- and intra-species variation in the cardiomyocyte composition of the adult heart, and may be relevant to understanding heart regenerative ability in humans and across other species.
Collapse
|
26
|
Cerrone M, Remme CA, Tadros R, Bezzina CR, Delmar M. Beyond the One Gene-One Disease Paradigm: Complex Genetics and Pleiotropy in Inheritable Cardiac Disorders. Circulation 2019; 140:595-610. [PMID: 31403841 DOI: 10.1161/circulationaha.118.035954] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inheritable cardiac disorders, which may be associated with cardiomyopathic changes, are often associated with increased risk of sudden death in the young. Early linkage analysis studies in Mendelian forms of these diseases, such as hypertrophic cardiomyopathy and long-QT syndrome, uncovered large-effect genetic variants that contribute to the phenotype. In more recent years, through genotype-phenotype studies and methodological advances in genetics, it has become evident that most inheritable cardiac disorders are not monogenic but, rather, have a complex genetic basis wherein multiple genetic variants contribute (oligogenic or polygenic inheritance). Conversely, studies on genes underlying these disorders uncovered pleiotropic effects, with a single gene affecting multiple and apparently unrelated phenotypes. In this review, we explore these 2 phenomena: on the one hand, the evidence that variants in multiple genes converge to generate one clinical phenotype, and, on the other, the evidence that variants in one gene can lead to apparently unrelated phenotypes. Although multiple conditions are addressed to illustrate these concepts, the experience obtained in the study of long-QT syndrome, Brugada syndrome, and arrhythmogenic cardiomyopathy, and in the study of functions related to SCN5A (the gene coding for the α-subunit of the most abundant sodium channel in the heart) and PKP2 (the gene coding for the desmosomal protein plakophilin-2), as well, is discussed in more detail.
Collapse
Affiliation(s)
- Marina Cerrone
- Leon H. Charney Division of Cardiology (M.C., M.D.), NYU School of Medicine, New York.,Inherited Arrhythmias Clinic and Heart Rhythm Center, Leon H. Charney Division of Cardiology (M.C.), NYU School of Medicine, New York
| | - Carol Ann Remme
- Inherited Arrhythmias Clinic and Heart Rhythm Center, Leon H. Charney Division of Cardiology (M.C.), NYU School of Medicine, New York
| | - Rafik Tadros
- Amsterdam UMC, University of Amsterdam, Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, AMC Heart Center, The Netherlands (C.A.R., C.R.B.)
| | - Connie R Bezzina
- Inherited Arrhythmias Clinic and Heart Rhythm Center, Leon H. Charney Division of Cardiology (M.C.), NYU School of Medicine, New York
| | - Mario Delmar
- Leon H. Charney Division of Cardiology (M.C., M.D.), NYU School of Medicine, New York
| |
Collapse
|
27
|
Olivaes J, Bonamino MH, Markoski MM. CRISPR/Cas 9 system for the treatment of dilated cardiomyopathy: A hypothesis related to function of a MAP kinase. Med Hypotheses 2019; 128:91-93. [DOI: 10.1016/j.mehy.2019.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/23/2019] [Accepted: 05/12/2019] [Indexed: 10/26/2022]
|
28
|
da Costa Martins PA. Mononuclear Diploidy at the Heart of Cardiomyocyte Proliferation. Cell Stem Cell 2019; 21:421-422. [PMID: 28985522 DOI: 10.1016/j.stem.2017.09.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Reporting in Nature Genetics, Patterson et al. (2017) show that adult mammalian hearts possess an innate capacity to regenerate, which depends on a small population of mononuclear diploid cardiomyocytes. These cells undergo karyokinesis and cytokinesis, raising the possibility that endogenous cardiac muscle cells can be stimulated to proliferate for myocardial repair.
Collapse
Affiliation(s)
- Paula A da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
29
|
Dittmann S, Schulze-Bahr E. Nonsynonymous TNNI3K mutations and phenotypes: More than a "simple" cardiomyopathy. Heart Rhythm 2019; 16:106-107. [PMID: 30063992 DOI: 10.1016/j.hrthm.2018.07.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Sven Dittmann
- Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Münster, Germany
| | - Eric Schulze-Bahr
- Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Münster, Germany.
| |
Collapse
|
30
|
Podliesna S, Delanne J, Miller L, Tester DJ, Uzunyan M, Yano S, Klerk M, Cannon BC, Khongphatthanayothin A, Laurent G, Bertaux G, Falcon-Eicher S, Wu S, Yen HY, Gao H, Wilde AAM, Faivre L, Ackerman MJ, Lodder EM, Bezzina CR. Supraventricular tachycardias, conduction disease, and cardiomyopathy in 3 families with the same rare variant in TNNI3K (p.Glu768Lys). Heart Rhythm 2018; 16:98-105. [PMID: 30010057 DOI: 10.1016/j.hrthm.2018.07.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Rare genetic variants in TNNI3K encoding troponin-I interacting kinase have been linked to a distinct syndrome consisting primarily of supraventricular tachycardias and variably expressed conduction disturbance and dilated cardiomyopathy in 2 families. OBJECTIVE The purpose of this study was to identify new genetic variants associated with inherited supraventricular tachycardias, cardiac conduction disease, and cardiomyopathy. METHODS We conducted next generation sequencing in 3 independent multigenerational families with atrial/junctional tachycardia with or without conduction disturbance, dilated cardiomyopathy, and sudden death. We also assessed the effect of identified variant on protein autophosphorylation. RESULTS In this study, we uncovered the same ultra-rare genetic variant in TNNI3K (c.2302G>A, p.Glu768Lys), which co-segregated with disease features in all affected individuals (n = 23) from all 3 families. TNNI3K harboring the TNNI3K-p.Glu768Lys variant displayed enhanced kinase activity, in line with expectations from previous mouse studies that demonstrated increased conduction indices and procardiomyopathic effects with increased levels of Tnni3k. CONCLUSION This study corroborates further the causal link between rare genetic variation in TNNI3K and this distinct complex phenotype, and points to enhanced kinase activity of TNNI3K as the underlying pathobiological mechanism.
Collapse
Affiliation(s)
- Svitlana Podliesna
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Lindsey Miller
- USC Keck School of Medicine, LAC+USC Medical Center, Los Angeles, California
| | - David J Tester
- Departments of Cardiovascular Medicine (Division of Heart Rhythm Services), Pediatric and Adolescent Medicine (Division of Pediatric Cardiology), and Molecular Pharmacology & Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory), Mayo Clinic, Rochester, Minnesota
| | - Merujan Uzunyan
- USC Keck School of Medicine, LAC+USC Medical Center, Los Angeles, California
| | - Shoji Yano
- USC Keck School of Medicine, LAC+USC Medical Center, Los Angeles, California
| | - Mischa Klerk
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Bryan C Cannon
- Departments of Cardiovascular Medicine (Division of Heart Rhythm Services), Pediatric and Adolescent Medicine (Division of Pediatric Cardiology), and Molecular Pharmacology & Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory), Mayo Clinic, Rochester, Minnesota
| | - Apichai Khongphatthanayothin
- USC Keck School of Medicine, LAC+USC Medical Center, Los Angeles, California; Bangkok General Hospital and Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Gabriel Laurent
- Centre de compétence pour les troubles du rythme cardiaque d'origine génétique, CHU Dijon-Bourgogne, Dijon, France; Service de rythmologie Centre Hospitalier Universitaire Le Bocage 2, Dijon, France
| | - Geraldine Bertaux
- Centre de compétence pour les troubles du rythme cardiaque d'origine génétique, CHU Dijon-Bourgogne, Dijon, France
| | - Sylvie Falcon-Eicher
- Centre de compétence pour les troubles du rythme cardiaque d'origine génétique, CHU Dijon-Bourgogne, Dijon, France
| | | | | | - Hanlin Gao
- Fulgent Genetics, Temple City, California
| | - Arthur A M Wilde
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, Amsterdam, The Netherlands; Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders, Jeddah, Kingdom of Saudi Arabia
| | - Laurence Faivre
- Centre de Génétique, Hôpital d'Enfants, Dijon, France; Equipe GAD, UMR1231, FHU TRANSLAD et Institut GIMI, CHU Dijon-Bourgogne et Université de Bourgogne-Franche Comté, Dijon, France
| | - Michael J Ackerman
- Departments of Cardiovascular Medicine (Division of Heart Rhythm Services), Pediatric and Adolescent Medicine (Division of Pediatric Cardiology), and Molecular Pharmacology & Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory), Mayo Clinic, Rochester, Minnesota
| | - Elisabeth M Lodder
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
31
|
Philp J, Lawhorn BG, Graves AP, Shewchuk L, Rivera KL, Jolivette LJ, Holt DA, Gatto GJ, Kallander LS. 4,6-Diaminopyrimidines as Highly Preferred Troponin I-Interacting Kinase (TNNI3K) Inhibitors. J Med Chem 2018; 61:3076-3088. [PMID: 29561151 DOI: 10.1021/acs.jmedchem.8b00125] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Structure-guided progression of a purine-derived series of TNNI3K inhibitors directed design efforts that produced a novel series of 4,6-diaminopyrimidine inhibitors, an emerging kinase binding motif. Herein, we report a detailed understanding of the intrinsic conformational preferences of the scaffold, which impart high specificity for TNNI3K. Further manipulation of the template based on the conformational analysis and additional structure-activity relationship studies provided enhancements in kinase selectivity and pharmacokinetics that furnished an advanced series of potent inhibitors. The optimized compounds (e.g., GSK854) are suitable leads for identifying new cardiac medicines and have been employed as in vivo tools in investigational studies aimed at defining the role of TNNI3K within heart failure.
Collapse
|
32
|
Vujic A, Bassaneze V, Lee RT. Genetic insights into mammalian heart regeneration. Nat Genet 2017; 49:1292-1293. [PMID: 28854178 DOI: 10.1038/ng.3942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Genetic and functional analyses of 120 mouse strains have identified a heart regeneration candidate gene that modulates the contractile sarcomeric apparatus. This gene, Tnni3k, controls the frequency of the mononuclear, diploid cardiomyocyte population, which affects cardiomyocyte proliferative potential after injury.
Collapse
Affiliation(s)
- Ana Vujic
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Vinícius Bassaneze
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
33
|
Patterson M, Barske L, Van Handel B, Rau CD, Gan P, Sharma A, Parikh S, Denholtz M, Huang Y, Yamaguchi Y, Shen H, Allayee H, Crump JG, Force TI, Lien CL, Makita T, Lusis AJ, Kumar SR, Sucov HM. Frequency of mononuclear diploid cardiomyocytes underlies natural variation in heart regeneration. Nat Genet 2017; 49:1346-1353. [PMID: 28783163 DOI: 10.1038/ng.3929] [Citation(s) in RCA: 252] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/11/2017] [Indexed: 12/16/2022]
Abstract
Adult mammalian cardiomyocyte regeneration after injury is thought to be minimal. Mononuclear diploid cardiomyocytes (MNDCMs), a relatively small subpopulation in the adult heart, may account for the observed degree of regeneration, but this has not been tested. We surveyed 120 inbred mouse strains and found that the frequency of adult mononuclear cardiomyocytes was surprisingly variable (>7-fold). Cardiomyocyte proliferation and heart functional recovery after coronary artery ligation both correlated with pre-injury MNDCM content. Using genome-wide association, we identified Tnni3k as one gene that influences variation in this composition and demonstrated that Tnni3k knockout resulted in elevated MNDCM content and increased cardiomyocyte proliferation after injury. Reciprocally, overexpression of Tnni3k in zebrafish promoted cardiomyocyte polyploidization and compromised heart regeneration. Our results corroborate the relevance of MNDCMs in heart regeneration. Moreover, they imply that intrinsic heart regeneration is not limited nor uniform in all individuals, but rather is a variable trait influenced by multiple genes.
Collapse
Affiliation(s)
- Michaela Patterson
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Lindsey Barske
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ben Van Handel
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Christoph D Rau
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Peiheng Gan
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Avneesh Sharma
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Shan Parikh
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Matt Denholtz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Ying Huang
- Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Yukiko Yamaguchi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Hua Shen
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Hooman Allayee
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Thomas I Force
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ching-Ling Lien
- Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA.,Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Takako Makita
- Developmental Neuroscience Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Aldons J Lusis
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - S Ram Kumar
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Henry M Sucov
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
34
|
Kumar A, Rani B, Sharma R, Kaur G, Prasad R, Bahl A, Khullar M. ACE2, CALM3 and TNNI3K polymorphisms as potential disease modifiers in hypertrophic and dilated cardiomyopathies. Mol Cell Biochem 2017; 438:167-174. [PMID: 28744816 DOI: 10.1007/s11010-017-3123-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/15/2017] [Indexed: 12/18/2022]
Abstract
The marked clinical and genetic heterogeneity seen in hypertrophic (HCM) and dilated cardiomyopathies (DCM) suggests involvement of disease modifiers and environmental factors in the pathophysiology of these diseases. In the current study, we examined association of single nucleotide polymorphisms (SNPs) of three candidate genes, ACE2 (rs6632677), TNNI3K (rs49812611) and CALM3 (rs13477425) with clinical phenotypes of HCM and DCM patients of North Indian ethnicity. Prevalence of ACE2 (7160726 C>G) variant genotypes (CG and GG) was significantly higher in DCM subjects as compared to controls. Prevalence of TNNI3K (3784 C>T) and CALM3 (-34T>A) variant homozygous genotype were significantly higher in HCM and DCM subjects as compared to controls. DCM patients with CT genotype showed significant decrease in LVEF as compared to CC genotype (p < 0.03). There was significant gene-gene interaction between these SNPs and three-way SNP combination of ACE2 C>G, TNN13K C>T, CALM3 A>T gene variants and was associated with high risk of HCM and DCM. Presence of ACE2 (7160726 C>G) and CALM3 (-34T>A) variant genotypes in HCM Patients with mutations (sarcomeric or non sarcomeric genes) was associated with increased mean septal thickness, further suggesting a role of these gene variants in modifying disease phenotype. Our results suggest that ACE2, TNNI3K and CALM3 polymorphisms are associated with increased risk of HCM and DCM and may act as disease modifiers of these diseases.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Experimental Medicine and Biotechnology, PGIMER, Lab No 2009, Research Block B, Chandigarh, 160012, India
| | - Bindu Rani
- Department of Experimental Medicine and Biotechnology, PGIMER, Lab No 2009, Research Block B, Chandigarh, 160012, India
| | - Rajni Sharma
- Department of Otolaryngology, PGIMER, Chandigarh, India
| | - Gurjeet Kaur
- Department of Endocrinology, PGIMER, Chandigarh, India
| | - Rishikesh Prasad
- Department of Experimental Medicine and Biotechnology, PGIMER, Lab No 2009, Research Block B, Chandigarh, 160012, India
| | - Ajay Bahl
- Department of Cardiology, PGIMER, Chandigarh, India
| | - Madhu Khullar
- Department of Experimental Medicine and Biotechnology, PGIMER, Lab No 2009, Research Block B, Chandigarh, 160012, India.
| |
Collapse
|
35
|
Rau CD, Romay MC, Tuteryan M, Wang JJC, Santolini M, Ren S, Karma A, Weiss JN, Wang Y, Lusis AJ. Systems Genetics Approach Identifies Gene Pathways and Adamts2 as Drivers of Isoproterenol-Induced Cardiac Hypertrophy and Cardiomyopathy in Mice. Cell Syst 2017; 4:121-128.e4. [PMID: 27866946 PMCID: PMC5338604 DOI: 10.1016/j.cels.2016.10.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 09/09/2016] [Accepted: 10/19/2016] [Indexed: 10/20/2022]
Abstract
We previously reported a genetic analysis of heart failure traits in a population of inbred mouse strains treated with isoproterenol to mimic catecholamine-driven cardiac hypertrophy. Here, we apply a co-expression network algorithm, wMICA, to perform a systems-level analysis of left ventricular transcriptomes from these mice. We describe the features of the overall network but focus on a module identified in treated hearts that is strongly related to cardiac hypertrophy and pathological remodeling. Using the causal modeling algorithm NEO, we identified the gene Adamts2 as a putative regulator of this module and validated the predictive value of NEO using small interfering RNA-mediated knockdown in neonatal rat ventricular myocytes. Adamts2 silencing regulated the expression of the genes residing within the module and impaired isoproterenol-induced cellular hypertrophy. Our results provide a view of higher order interactions in heart failure with potential for diagnostic and therapeutic insights.
Collapse
Affiliation(s)
- Christoph D Rau
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Milagros C Romay
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mary Tuteryan
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jessica J-C Wang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marc Santolini
- Center for Interdisciplinary Research on Complex Systems, Department of Physics, Northeastern University, Boston, MA 02115, USA
| | - Shuxun Ren
- Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alain Karma
- Center for Interdisciplinary Research on Complex Systems, Department of Physics, Northeastern University, Boston, MA 02115, USA
| | - James N Weiss
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yibin Wang
- Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aldons J Lusis
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
36
|
Arend MC, Pereira JO, Markoski MM. The CRISPR/Cas9 System and the Possibility of Genomic Edition for Cardiology. Arq Bras Cardiol 2017; 108:81-83. [PMID: 28146210 PMCID: PMC5245852 DOI: 10.5935/abc.20160200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 10/13/2016] [Indexed: 11/20/2022] Open
Affiliation(s)
- Marcela Corso Arend
- Laboratório de Cardiologia Molecular e Celular, Instituto de Cardiologia, Fundação Universitária de Cardiologia, Porto Alegre, RS, Brazil
| | - Jessica Olivaes Pereira
- Laboratório de Cardiologia Molecular e Celular, Instituto de Cardiologia, Fundação Universitária de Cardiologia, Porto Alegre, RS, Brazil
| | - Melissa Medeiros Markoski
- Laboratório de Cardiologia Molecular e Celular, Instituto de Cardiologia, Fundação Universitária de Cardiologia, Porto Alegre, RS, Brazil
| |
Collapse
|
37
|
Lawhorn BG, Philp J, Graves AP, Holt DA, Gatto GJ, Kallander LS. Substituent Effects on Drug–Receptor H-bond Interactions: Correlations Useful for the Design of Kinase Inhibitors. J Med Chem 2016; 59:10629-10641. [DOI: 10.1021/acs.jmedchem.6b01342] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Brian G. Lawhorn
- Heart Failure Discovery Performance Unit and ‡Platform Technology
and Sciences, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Joanne Philp
- Heart Failure Discovery Performance Unit and ‡Platform Technology
and Sciences, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Alan P. Graves
- Heart Failure Discovery Performance Unit and ‡Platform Technology
and Sciences, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Dennis A. Holt
- Heart Failure Discovery Performance Unit and ‡Platform Technology
and Sciences, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Gregory J. Gatto
- Heart Failure Discovery Performance Unit and ‡Platform Technology
and Sciences, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Lara S. Kallander
- Heart Failure Discovery Performance Unit and ‡Platform Technology
and Sciences, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| |
Collapse
|
38
|
Ding Y, Long PA, Bos JM, Shih YH, Ma X, Sundsbak RS, Chen J, Jiang Y, Zhao L, Hu X, Wang J, Shi Y, Ackerman MJ, Lin X, Ekker SC, Redfield MM, Olson TM, Xu X. A modifier screen identifies DNAJB6 as a cardiomyopathy susceptibility gene. JCI Insight 2016; 1. [PMID: 27642634 DOI: 10.1172/jci.insight.88797] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Mutagenesis screening is a powerful forward genetic approach that has been successfully applied in lower-model organisms to discover genetic factors for biological processes. This phenotype-based approach has yet to be established in vertebrates for probing major human diseases, largely because of the complexity of colony management. Herein, we report a rapid strategy for identifying genetic modifiers of cardiomyopathy (CM). Based on the application of doxorubicin stress to zebrafish insertional cardiac (ZIC) mutants, we identified 4 candidate CM-modifying genes, of which 3 have been linked previously to CM. The long isoform of DnaJ (Hsp40) homolog, subfamily B, member 6b (dnajb6b(L)) was identified as a CM susceptibility gene, supported by identification of rare variants in its human ortholog DNAJB6 from CM patients. Mechanistic studies indicated that the deleterious, loss-of-function modifying effects of dnajb6b(L) can be ameliorated by inhibition of ER stress. In contrast, overexpression of dnajb6(L) exerts cardioprotective effects on both fish and mouse CM models. Together, our findings establish a mutagenesis screening strategy that is scalable for systematic identification of genetic modifiers of CM, feasible to suggest therapeutic targets, and expandable to other major human diseases.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA; Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Pamela A Long
- Cardiovascular Genetics Research Laboratory, Mayo Clinic, Rochester, Minnesota, USA
| | - J Martijn Bos
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Yu-Huan Shih
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA; Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Xiao Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA; Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Rhianna S Sundsbak
- Cardiovascular Genetics Research Laboratory, Mayo Clinic, Rochester, Minnesota, USA
| | - Jianhua Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Yiwen Jiang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Liqun Zhao
- Department of Cardiology, Shanghai First People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China
| | - Jianan Wang
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China
| | - Yongyong Shi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA; Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Michael J Ackerman
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA; Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Timothy M Olson
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA; Cardiovascular Genetics Research Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA; Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
39
|
Jeng XJ, Daye ZJ, Lu W, Tzeng JY. Rare Variants Association Analysis in Large-Scale Sequencing Studies at the Single Locus Level. PLoS Comput Biol 2016; 12:e1004993. [PMID: 27355347 PMCID: PMC4927097 DOI: 10.1371/journal.pcbi.1004993] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/21/2016] [Indexed: 11/24/2022] Open
Abstract
Genetic association analyses of rare variants in next-generation sequencing (NGS) studies are fundamentally challenging due to the presence of a very large number of candidate variants at extremely low minor allele frequencies. Recent developments often focus on pooling multiple variants to provide association analysis at the gene instead of the locus level. Nonetheless, pinpointing individual variants is a critical goal for genomic researches as such information can facilitate the precise delineation of molecular mechanisms and functions of genetic factors on diseases. Due to the extreme rarity of mutations and high-dimensionality, significances of causal variants cannot easily stand out from those of noncausal ones. Consequently, standard false-positive control procedures, such as the Bonferroni and false discovery rate (FDR), are often impractical to apply, as a majority of the causal variants can only be identified along with a few but unknown number of noncausal variants. To provide informative analysis of individual variants in large-scale sequencing studies, we propose the Adaptive False-Negative Control (AFNC) procedure that can include a large proportion of causal variants with high confidence by introducing a novel statistical inquiry to determine those variants that can be confidently dispatched as noncausal. The AFNC provides a general framework that can accommodate for a variety of models and significance tests. The procedure is computationally efficient and can adapt to the underlying proportion of causal variants and quality of significance rankings. Extensive simulation studies across a plethora of scenarios demonstrate that the AFNC is advantageous for identifying individual rare variants, whereas the Bonferroni and FDR are exceedingly over-conservative for rare variants association studies. In the analyses of the CoLaus dataset, AFNC has identified individual variants most responsible for gene-level significances. Moreover, single-variant results using the AFNC have been successfully applied to infer related genes with annotation information. Next-generation sequencing technologies have allowed genetic association studies of complex traits at the single base-pair resolution, where most genetic variants have extremely low mutation frequencies. These rare variants have been the focus of modern statistical-computational genomics due to their potential to explain missing disease heritability. The identification of individual rare variants associated with diseases can provide new biological insights and enable the precise delineation of disease mechanisms. However, due to the extreme rarity of mutations and large numbers of variants, significances of causative variants tend to be mixed inseparably with a few noncausative ones, and standard multiple testing procedures controlling for false positives fail to provide a meaningful way to include a large proportion of the causative variants. To address the challenge of detecting weak biological signals, we propose a novel statistical procedure, based on false-negative control, to provide a practical approach for variant inclusion in large-scale sequencing studies. By determining those variants that can be confidently dispatched as noncausative, the proposed procedure offers an objective selection of a modest number of potentially causative variants at the single-locus level. Results can be further prioritized or used to infer disease-associated genes with annotation information.
Collapse
Affiliation(s)
- Xinge Jessie Jeng
- Department of Statistics, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Zhongyin John Daye
- Epidemiology and Biostatistics, University of Arizona, Tucson, Arizona, United States of America
| | - Wenbin Lu
- Department of Statistics, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Jung-Ying Tzeng
- Department of Statistics, North Carolina State University, Raleigh, North Carolina, United States of America
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Statistics, National Cheng-Kung University, Tainan, Taiwan
- * E-mail:
| |
Collapse
|
40
|
Lawhorn BG, Philp J, Graves AP, Shewchuk L, Holt DA, Gatto GJ, Kallander LS. GSK114: A selective inhibitor for elucidating the biological role of TNNI3K. Bioorg Med Chem Lett 2016; 26:3355-3358. [PMID: 27246618 DOI: 10.1016/j.bmcl.2016.05.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 12/31/2022]
Abstract
A series of selective TNNI3K inhibitors were developed by modifying the hinge-binding heterocycle of a previously reported dual TNNI3K/B-Raf inhibitor. The resulting quinazoline-containing compounds exhibit a large preference (up to 250-fold) for binding to TNNI3K versus B-Raf, are useful probes for elucidating the biological pathways associated with TNNI3K, and are leads for discovering novel cardiac medicines. GSK114 emerged as a leading inhibitor, displaying significant bias (40-fold) for TNNI3K over B-Raf, exceptional broad spectrum kinase selectivity, and adequate oral exposure to enable its use in cellular and in vivo studies.
Collapse
Affiliation(s)
- Brian G Lawhorn
- Heart Failure DPU, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, PA 19406, USA.
| | - Joanne Philp
- Heart Failure DPU, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, PA 19406, USA
| | - Alan P Graves
- Platform Sciences and Technology, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, PA 19406, USA
| | - Lisa Shewchuk
- Platform Sciences and Technology, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, PA 19406, USA
| | - Dennis A Holt
- Heart Failure DPU, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, PA 19406, USA
| | - Gregory J Gatto
- Heart Failure DPU, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, PA 19406, USA
| | - Lara S Kallander
- Heart Failure DPU, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, PA 19406, USA
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW In contrast to many other human diseases, the use of genome-wide association studies (GWAS) to identify genes for heart failure (HF) has had limited success. We will discuss the underlying challenges as well as potential new approaches to understanding the genetics of common forms of HF. RECENT FINDINGS Recent research using intermediate phenotypes, more detailed and quantitative stratification of HF symptoms, founder populations and novel animal models has begun to allow researchers to make headway toward explaining the genetics underlying HF using GWAS techniques. SUMMARY By expanding analyses of HF to improved clinical traits, additional HF classifications and innovative model systems, the intractability of human HF GWAS should be ameliorated significantly.
Collapse
|
42
|
Lawhorn BG, Philp J, Zhao Y, Louer C, Hammond M, Cheung M, Fries H, Graves AP, Shewchuk L, Wang L, Cottom JE, Qi H, Zhao H, Totoritis R, Zhang G, Schwartz B, Li H, Sweitzer S, Holt DA, Gatto GJ, Kallander LS. Identification of Purines and 7-Deazapurines as Potent and Selective Type I Inhibitors of Troponin I-Interacting Kinase (TNNI3K). J Med Chem 2015; 58:7431-48. [PMID: 26355916 DOI: 10.1021/acs.jmedchem.5b00931] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
A series of cardiac troponin I-interacting kinase (TNNI3K) inhibitors arising from 3-((9H-purin-6-yl)amino)-N-methyl-benzenesulfonamide (1) is disclosed along with fundamental structure-function relationships that delineate the role of each element of 1 for TNNI3K recognition. An X-ray structure of 1 bound to TNNI3K confirmed its Type I binding mode and is used to rationalize the structure-activity relationship and employed to design potent, selective, and orally bioavailable TNNI3K inhibitors. Identification of the 7-deazapurine heterocycle as a superior template (vs purine) and its elaboration by introduction of C4-benzenesulfonamide and C7- and C8-7-deazapurine substituents produced compounds with substantial improvements in potency (>1000-fold), general kinase selectivity (10-fold improvement), and pharmacokinetic properties (>10-fold increase in poDNAUC). Optimal members of the series have properties suitable for use in in vitro and in vivo experiments aimed at elucidating the role of TNNI3K in cardiac biology and serve as leads for developing novel heart failure medicines.
Collapse
Affiliation(s)
- Brian G Lawhorn
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Joanne Philp
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Yongdong Zhao
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Christopher Louer
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Marlys Hammond
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Mui Cheung
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Harvey Fries
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Alan P Graves
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Lisa Shewchuk
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Liping Wang
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Joshua E Cottom
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Hongwei Qi
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Huizhen Zhao
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Rachel Totoritis
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Guofeng Zhang
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Benjamin Schwartz
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Hu Li
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Sharon Sweitzer
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Dennis A Holt
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Gregory J Gatto
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Lara S Kallander
- Heart Failure Discovery Performance Unit and ‡Platform Technology and Sciences, GlaxoSmithKline , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| |
Collapse
|
43
|
Kimura A. Molecular genetics and pathogenesis of cardiomyopathy. J Hum Genet 2015; 61:41-50. [PMID: 26178429 DOI: 10.1038/jhg.2015.83] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 06/15/2015] [Indexed: 12/19/2022]
Abstract
Cardiomyopathy is defined as a disease of functional impairment in the cardiac muscle and its etiology includes both extrinsic and intrinsic factors. Cardiomyopathy caused by the intrinsic factors is called as primary cardiomyopathy of which two major clinical phenotypes are hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). Genetic approaches have revealed the disease genes for hereditary primary cardiomyopathy and functional studies have demonstrated that characteristic functional alterations induced by the disease-associated mutations are closely related to the clinical types, such that increased and decreased Ca(2+) sensitivities of muscle contraction are associated with HCM and DCM, respectively. In addition, recent studies have suggested that mutations in the Z-disc components found in HCM and DCM may result in increased and decreased stiffness of sarcomere, respectively. Moreover, functional analysis of mutations in the other components of cardiac muscle have suggested that the altered response to metabolic stresses is associated with cardiomyopathy, further indicating the heterogeneity in the etiology and pathogenesis of cardiomyopathy.
Collapse
Affiliation(s)
- Akinori Kimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
44
|
Milano A, Lodder EM, Bezzina CR. TNNI3K in cardiovascular disease and prospects for therapy. J Mol Cell Cardiol 2015; 82:167-73. [PMID: 25787061 DOI: 10.1016/j.yjmcc.2015.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 02/23/2015] [Accepted: 03/09/2015] [Indexed: 12/26/2022]
Abstract
Cardiovascular diseases are an important cause of morbidity and mortality worldwide and the global burden of these diseases continues to grow. Therefore new therapies are urgently needed. The role of protein kinases in disease, including cardiac disease, is long recognized, making kinases important therapeutic targets. We here review the knowledge gathered in the last decade about troponin I-interacting kinase (TNNI3K), a kinase with cardiac-restricted expression that has been implicated in various cardiac phenotypes and diseases including heart failure, cardiomyopathy, ischemia/reperfusion injury and conduction of the cardiac electrical impulse.
Collapse
Affiliation(s)
- Annalisa Milano
- Department of Clinical and Experimental Cardiology, Academic Medical Centre, Amsterdam, The Netherlands
| | - Elisabeth M Lodder
- Department of Clinical and Experimental Cardiology, Academic Medical Centre, Amsterdam, The Netherlands
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Academic Medical Centre, Amsterdam, The Netherlands.
| |
Collapse
|
45
|
Xi Y, Honeywell C, Zhang D, Schwartzentruber J, Beaulieu CL, Tetreault M, Hartley T, Marton J, Vidal SM, Majewski J, Aravind L, Gollob M, Boycott KM, Gow RM. Whole exome sequencing identifies the TNNI3K gene as a cause of familial conduction system disease and congenital junctional ectopic tachycardia. Int J Cardiol 2015; 185:114-6. [PMID: 25791106 DOI: 10.1016/j.ijcard.2015.03.130] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 03/07/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Yanwei Xi
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Christina Honeywell
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Dapeng Zhang
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | | | - Chandree L Beaulieu
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Martine Tetreault
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada; Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Taila Hartley
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Jennifer Marton
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Silvia M Vidal
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Jacek Majewski
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada; Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - L Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | | | - Michael Gollob
- Toronto General Hospital, Department of Cardiology, University of Toronto, Toronto, Ontario, Canada
| | - Kym M Boycott
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada.
| | - Robert M Gow
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada.
| |
Collapse
|
46
|
Abstract
Heart failure is highly influenced by heritability, and nearly 100 genes link to familial cardiomyopathy. Despite the marked genetic diversity that underlies these complex cardiovascular phenotypes, several key genes and pathways have emerged. Hypertrophic cardiomyopathy is characterized by increased contractility and a greater energetic cost of cardiac output. Dilated cardiomyopathy is often triggered by mutations that disrupt the giant protein titin. The energetic consequences of these mutations offer molecular targets and opportunities for new drug development and gene correction therapies.
Collapse
Affiliation(s)
- Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - David Y Barefield
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Megan J Puckelwartz
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
47
|
Rau CD, Wang J, Avetisyan R, Romay MC, Martin L, Ren S, Wang Y, Lusis AJ. Mapping genetic contributions to cardiac pathology induced by Beta-adrenergic stimulation in mice. ACTA ACUST UNITED AC 2014; 8:40-9. [PMID: 25480693 DOI: 10.1161/circgenetics.113.000732] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chronic stress-induced cardiac pathology exhibits both a wide range in severity and a high degree of heterogeneity in clinical manifestation in human patients. This variability is contributed to by complex genetic and environmental etiologies within the human population. Genetic approaches to elucidate the genetics underlying the acquired forms of cardiomyopathies, including genome-wide association studies, have been largely unsuccessful, resulting in limited knowledge as to the contribution of genetic variations for this important disease. METHODS AND RESULTS Using the β-adrenergic agonist isoproterenol as a specific pathological stressor to circumvent the problem of etiologic heterogeneity, we performed a genome-wide association study for genes influencing cardiac hypertrophy and fibrosis in a large panel of inbred mice. Our analyses revealed 7 significant loci and 17 suggestive loci, containing an average of 14 genes, affecting cardiac hypertrophy, fibrosis, and surrogate traits relevant to heart failure. Several loci contained candidate genes which are known to contribute to Mendelian cardiomyopathies in humans or have established roles in cardiac pathology based on molecular or genetic studies in mouse models. In particular, we identify Abcc6 as a novel gene underlying a fibrosis locus by validating that an allele with a splice mutation of Abcc6 dramatically and rapidly promotes isoproterenol-induced cardiac fibrosis. CONCLUSIONS Genetic variants significantly contribute to the phenotypic heterogeneity of stress-induced cardiomyopathy. Systems genetics is an effective approach to identify genes and pathways underlying the specific pathological features of cardiomyopathies. Abcc6 is a previously unrecognized player in the development of stress-induced cardiac fibrosis.
Collapse
Affiliation(s)
- Christoph D Rau
- From the Department of Microbiology, Immunology and Molecular Genetics, Department of Human Genetics (C.D.R., R.A., M.R., A.J.L.), Department of Medicine, Division of Cardiology, David Geffen School of Medicine (J.W., L.M., A.J.L.), and Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine (S.R., Y.W.), University of California, Los Angeles, CA
| | - Jessica Wang
- From the Department of Microbiology, Immunology and Molecular Genetics, Department of Human Genetics (C.D.R., R.A., M.R., A.J.L.), Department of Medicine, Division of Cardiology, David Geffen School of Medicine (J.W., L.M., A.J.L.), and Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine (S.R., Y.W.), University of California, Los Angeles, CA
| | - Rozeta Avetisyan
- From the Department of Microbiology, Immunology and Molecular Genetics, Department of Human Genetics (C.D.R., R.A., M.R., A.J.L.), Department of Medicine, Division of Cardiology, David Geffen School of Medicine (J.W., L.M., A.J.L.), and Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine (S.R., Y.W.), University of California, Los Angeles, CA
| | - Milagros C Romay
- From the Department of Microbiology, Immunology and Molecular Genetics, Department of Human Genetics (C.D.R., R.A., M.R., A.J.L.), Department of Medicine, Division of Cardiology, David Geffen School of Medicine (J.W., L.M., A.J.L.), and Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine (S.R., Y.W.), University of California, Los Angeles, CA
| | - Lisa Martin
- From the Department of Microbiology, Immunology and Molecular Genetics, Department of Human Genetics (C.D.R., R.A., M.R., A.J.L.), Department of Medicine, Division of Cardiology, David Geffen School of Medicine (J.W., L.M., A.J.L.), and Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine (S.R., Y.W.), University of California, Los Angeles, CA
| | - Shuxun Ren
- From the Department of Microbiology, Immunology and Molecular Genetics, Department of Human Genetics (C.D.R., R.A., M.R., A.J.L.), Department of Medicine, Division of Cardiology, David Geffen School of Medicine (J.W., L.M., A.J.L.), and Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine (S.R., Y.W.), University of California, Los Angeles, CA
| | - Yibin Wang
- From the Department of Microbiology, Immunology and Molecular Genetics, Department of Human Genetics (C.D.R., R.A., M.R., A.J.L.), Department of Medicine, Division of Cardiology, David Geffen School of Medicine (J.W., L.M., A.J.L.), and Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine (S.R., Y.W.), University of California, Los Angeles, CA.
| | - Aldons J Lusis
- From the Department of Microbiology, Immunology and Molecular Genetics, Department of Human Genetics (C.D.R., R.A., M.R., A.J.L.), Department of Medicine, Division of Cardiology, David Geffen School of Medicine (J.W., L.M., A.J.L.), and Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine (S.R., Y.W.), University of California, Los Angeles, CA.
| |
Collapse
|
48
|
Abraham DM, Marchuk DA. Inhibition of the cardiomyocyte-specific troponin I-interacting kinase limits oxidative stress, injury, and adverse remodeling due to ischemic heart disease. Circ Res 2014; 114:938-40. [PMID: 24625723 DOI: 10.1161/circresaha.113.303238] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ischemia–reperfusion injury is strongly associated with increased oxidative stress, mitochondrial dysfunction, and cell death. These processes are diminished in an animal model of ischemia–reperfusion by the genetic loss or pharmacological inhibition of troponin I–interacting kinase.
Collapse
Affiliation(s)
- Dennis M Abraham
- From the Department of Medicine, Division of Cardiology (D.M.A.) and Department of Molecular Genetics and Microbiology (D.A.M.), Duke University School of Medicine, Durham, NC
| | | |
Collapse
|
49
|
Vagnozzi RJ, Gatto GJ, Kallander LS, Hoffman NE, Mallilankaraman K, Ballard VLT, Lawhorn BG, Stoy P, Philp J, Graves AP, Naito Y, Lepore JJ, Gao E, Madesh M, Force T. Inhibition of the cardiomyocyte-specific kinase TNNI3K limits oxidative stress, injury, and adverse remodeling in the ischemic heart. Sci Transl Med 2014; 5:207ra141. [PMID: 24132636 DOI: 10.1126/scitranslmed.3006479] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Percutaneous coronary intervention is first-line therapy for acute coronary syndromes (ACS) but can promote cardiomyocyte death and cardiac dysfunction via reperfusion injury, a phenomenon driven in large part by oxidative stress. Therapies to limit this progression have proven elusive, with no major classes of new agents since the development of anti-platelets/anti-thrombotics. We report that cardiac troponin I-interacting kinase (TNNI3K), a cardiomyocyte-specific kinase, promotes ischemia/reperfusion injury, oxidative stress, and myocyte death. TNNI3K-mediated injury occurs through increased mitochondrial superoxide production and impaired mitochondrial function and is largely dependent on p38 mitogen-activated protein kinase (MAPK) activation. We developed a series of small-molecule TNNI3K inhibitors that reduce mitochondrial-derived superoxide generation, p38 activation, and infarct size when delivered at reperfusion to mimic clinical intervention. TNNI3K inhibition also preserves cardiac function and limits chronic adverse remodeling. Our findings demonstrate that TNNI3K modulates reperfusion injury in the ischemic heart and is a tractable therapeutic target for ACS. Pharmacologic TNNI3K inhibition would be cardiac-selective, preventing potential adverse effects of systemic kinase inhibition.
Collapse
Affiliation(s)
- Ronald J Vagnozzi
- Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Theis JL, Zimmermann MT, Larsen BT, Rybakova IN, Long PA, Evans JM, Middha S, de Andrade M, Moss RL, Wieben ED, Michels VV, Olson TM. TNNI3K mutation in familial syndrome of conduction system disease, atrial tachyarrhythmia and dilated cardiomyopathy. Hum Mol Genet 2014; 23:5793-804. [PMID: 24925317 DOI: 10.1093/hmg/ddu297] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Locus mapping has uncovered diverse etiologies for familial atrial fibrillation (AF), dilated cardiomyopathy (DCM), and mixed cardiac phenotype syndromes, yet the molecular basis for these disorders remains idiopathic in most cases. Whole-exome sequencing (WES) provides a powerful new tool for familial disease gene discovery. Here, synergistic application of these genomic strategies identified the pathogenic mutation in a familial syndrome of atrial tachyarrhythmia, conduction system disease (CSD), and DCM vulnerability. Seven members of a three-generation family exhibited the variably expressed phenotype, three of whom manifested CSD and clinically significant arrhythmia in childhood. Genome-wide linkage analysis mapped two equally plausible loci to chromosomes 1p3 and 13q12. Variants from WES of two affected cousins were filtered for rare, predicted-deleterious, positional variants, revealing an unreported heterozygous missense mutation disrupting the highly conserved kinase domain in TNNI3K. The G526D substitution in troponin I interacting kinase, with the most deleterious SIFT and Polyphen2 scores possible, resulted in abnormal peptide aggregation in vitro and in silico docking models predicted altered yet energetically favorable wild-type mutant dimerization. Ventricular tissue from a mutation carrier displayed histopathological hallmarks of DCM and reduced TNNI3K protein staining with unique amorphous nuclear and sarcoplasmic inclusions. In conclusion, mutation of TNNI3K, encoding a heart-specific kinase previously shown to modulate cardiac conduction and myocardial function in mice, underlies a familial syndrome of electrical and myopathic heart disease. The identified substitution causes a TNNI3K aggregation defect and protein deficiency, implicating a dominant-negative loss of function disease mechanism.
Collapse
Affiliation(s)
| | - Michael T Zimmermann
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research
| | | | - Inna N Rybakova
- Department of Cell and Regenerative Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Jared M Evans
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research
| | - Sumit Middha
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research
| | - Mariza de Andrade
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research
| | - Richard L Moss
- Department of Cell and Regenerative Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | | | - Timothy M Olson
- Cardiovascular Genetics Research Laboratory, Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine,
| |
Collapse
|