Chromatin Compartment Dynamics in a Haploinsufficient Model of Cardiac Laminopathy

Total Page:16

File Type:pdf, Size:1020Kb

Chromatin Compartment Dynamics in a Haploinsufficient Model of Cardiac Laminopathy Published Online: 8 August, 2019 | Supp Info: http://doi.org/10.1083/jcb.201902117 Downloaded from jcb.rupress.org on August 14, 2019 ARTICLE Chromatin compartment dynamics in a haploinsufficient model of cardiac laminopathy Alessandro Bertero1,2,3,PaulA.Fields1,2,3, Alec S.T. Smith2,3,4, Andrea Leonard2,3,5, Kevin Beussman2,3,5, Nathan J. Sniadecki2,3,4,5, Deok-Ho Kim2,3,4, Hung-Fat Tse6, Lil Pabon1,2,3, Jay Shendure7,8, William S. Noble7, and Charles E. Murry1,2,3,4,9 Mutations in A-type nuclear lamins cause dilated cardiomyopathy, which is postulated to result from dysregulated gene expression due to changes in chromatin organization into active and inactive compartments. To test this, we performed genome-wide chromosome conformation analyses in human induced pluripotent stem cell–derived cardiomyocytes (hiPSC- CMs) with a haploinsufficient mutation for lamin A/C. Compared with gene-corrected cells, mutant hiPSC-CMs have marked electrophysiological and contractile alterations, with modest gene expression changes. While large-scale changes in chromosomal topology are evident, differences in chromatin compartmentalization are limited to a few hotspots that escape segregation to the nuclear lamina and inactivation during cardiogenesis. These regions exhibit up-regulation of multiple noncardiac genes including CACNA1A, encoding for neuronal P/Q-type calcium channels. Pharmacological inhibition of the resulting current partially mitigates the electrical alterations. However, chromatin compartment changes do not explain most gene expression alterations in mutant hiPSC-CMs. Thus, global errors in chromosomal compartmentation are not the primary pathogenic mechanism in heart failure due to lamin A/C haploinsufficiency. Introduction Chromatin organization in the 3D space has emerged as a key Fraser et al., 2015; Schmitt et al., 2016; Won et al., 2016; Bertero layer of mammalian gene expression control. The development et al., 2019). However, these same studies have established that of powerful technologies to map chromatin architecture, in pluripotent stem cell differentiation leads to a substantial degree particular genome-wide chromosome conformation capture (Hi- of A/B compartment reorganization (e.g., ∼20% of the genome), C; Lieberman-Aiden et al., 2009), has revealed that such 3D and that this is associated with important developmental structure is complex, nonrandom, and hierarchical (Yu and Ren, changes in gene expression. In the context of disease, disruption 2017). Short- and long-range intra-chromosomal (cis) DNA in- of TADs due to copy number variations or point mutations leads teractions are generally constrained within topologically asso- to congenital developmental disorders and cancers (Katainen ciating domains (TADs; Dixon et al., 2012; Nora et al., 2012; et al., 2015; Lupiañez´ et al., 2015; Redin et al., 2017; Sun et al., Sexton et al., 2012). TADs tend to interact based on their epi- 2018). In contrast, whether dysregulation of A/B compartments genetic status and transcriptional activity, thus dividing chro- plays a role in functional changes of gene expression leading to mosomes into two types of large-scale compartments generally human disease is still unclear (Krumm and Duan, 2018). called active and inactive (A and B, respectively; Simonis et al., The nuclear lamina has been proposed as a regulator of 2006; Lieberman-Aiden et al., 2009; Rao et al., 2014). chromatin compartmentalization in development and disease Moving beyond the third dimension, there has been growing (Buchwalter et al., 2019). The lamina lies along the inner nuclear interest in understanding the functional changes in chromatin membrane and is a complex mesh of nuclear intermediate fila- organization during development and in disease (Dekker et al., ments (A- and B-type lamins) and lamin-associated proteins. 2017). An emerging body of work has shown that TADs are This structure provides key mechanical support to the nucleus, largely invariant across cell types (Dixon et al., 2012, 2015; and is an important hub for the control of intracellular signaling ............................................................................................................................................................................. 1Department of Pathology, University of Washington, Seattle, WA; 2Center for Cardiovascular Biology, University of Washington, Seattle, WA; 3Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA; 4Department of Bioengineering, University of Washington, Seattle, WA; 5Department of Mechanical Engineering, University of Washington, Seattle, WA; 6Cardiology Division, Department of Medicine, University of Hong Kong, Pok Fu Lam, Hong Kong; 7Department of Genome Sciences, University of Washington, Seattle, WA; 8Howard Hughes Medical Institute, Seattle, WA; 9Department of Medicine/Cardiology, University of Washington, Seattle, WA. Correspondence to Charles E. Murry: [email protected]. © 2019 Bertero et al. This article is distributed under the terms of an Attribution–Noncommercial–Share Alike–No Mirror Sites license for the first six months after the publication date (see http://www.rupress.org/terms/). After six months it is available under a Creative Commons License (Attribution–Noncommercial–Share Alike 4.0 International license, as described at https://creativecommons.org/licenses/by-nc-sa/4.0/). Rockefeller University Press https://doi.org/10.1083/jcb.201902117 1 J. Cell Biol. 2019 (Dobrzynska et al., 2016). Moreover, the nuclear lamina inter- these studies have relied on fibroblasts instead of car- acts with large chromatin regions, aptly named lamin-associated diomyocytes, the primary cell type involved in cardiac lamin- domains (LADs; van Steensel and Belmont, 2017), which show opathy. Most importantly, to the best of our knowledge, the 3D heterochromatic features such as low gene density, enrichment chromatin organization changes associated with cardiac lam- for repressive histone marks, and poor transcriptional activity inopathy have not yet been tested at a genome-wide level. (Guelen et al., 2008). Several studies have established that such To address these limitations, we performed Hi-C and gene peripherally located LADs strongly correlate with the B com- expression (RNA sequencing [RNA-seq]) analyses to examine partment, while the A compartment is predominantly in the the changes in 3D chromatin architecture induced by a hap- nuclear interior (Kind et al., 2015; Luperchio et al., 2017; Stevens loinsufficient LMNA mutation in cardiomyocytes derived from et al., 2017; Zheng et al., 2018). Both lamins and lamin-associated human induced pluripotent stem cells (hiPSC-CMs). We hy- proteins, such as lamin B receptor, can directly interact with pothesized that decreased expression of A-type lamins would chromatin (Yuan et al., 1991; Ye and Worman, 1994, 1996; lead to broad functional alterations in A/B compartmentaliza- Taniura et al., 1995). Based on this and on the results of loss-of- tion, leading to aberrant gene expression. However, our findings function studies, it has been proposed that the nuclear lamina indicate that while lamin A/C haploinsufficiency functionally tethers LADs at the nuclear periphery (Solovei et al., 2013; Harr affects selected aspects of 3D chromatin organization in human et al., 2015; Luperchio et al., 2017). However, the precise role of cardiomyocytes, altered A/B compartmentalization does not A- and B-type lamins in chromatin compartmentalization is still represent the primary mechanism directly leading to gene ex- being debated (Amendola and van Steensel, 2015; Zheng et al., pression changes and disease pathogenesis. 2015, 2018; Adriaens et al., 2018). Elucidating the function of A-type lamins (lamin A and lamin C, which result from alternative splicing of the LMNA gene) is Results particularly important because of their involvement in human Generation of an in vitro model of cardiac lamin disease. LMNA mutations lead to a wide spectrum of conditions A/C haploinsufficiency collectively referred to as laminopathies, which most often affect To investigate the role of chromatin dynamics in cardiac lam- striated muscles (Capell and Collins, 2006; Bertrand et al., 2011). inopathy, we took advantage of hiPSCs bearing a heterozygous The majority of patients with striated muscle laminopathies nonsense mutation in LMNA predicted to cause premature develop dilated cardiomyopathy (DCM; Captur et al., 2018), and truncation of both lamin A and lamin C splicing isoforms mutations in LMNA are among the most common causes of fa- (c.672C>T, resulting in p.Arg225*, which we will refer to as milial DCM, depending on the ethnicity of the population R225X; Fig. 1 A). This hiPSC line was previously derived from a (Akinrinade et al., 2015; Haas et al., 2015; Tobita et al., 2018). 56-yr-old male patient who developed severe cardiac conduction Compared with other types of DCM, LMNA-DCM (which disease evolving into heart failure, a condition that segregated throughout the rest of the article we refer to as “cardiac lam- within the family with autosomal-dominant inheritance of the inopathy”) is quite atypical and more severe, as it is character- R225X mutation (Siu et al., 2012). This same mutation has been ized by early onset of life-threatening cardiac electrical reported in multiple other cohorts with similar symptoms abnormalities such as severe conduction system disease and/or (Jakobs et al., 2001; van Tintelen et al., 2007a; Saga et al., atrial and ventricular arrhythmias (van
Recommended publications
  • FLNC Pathogenic Variants in Patients with Cardiomyopathies
    FLNC pathogenic variants in patients with cardiomyopathies: Prevalence and genotype-phenotype correlations Flavie Ader, Pascal de Groote, Patricia Réant, Caroline Rooryck-Thambo, Delphine Dupin-Deguine, Caroline Rambaud, Diala Khraiche, Claire Perret, Jean-François Pruny, Michèle Mathieu-dramard, et al. To cite this version: Flavie Ader, Pascal de Groote, Patricia Réant, Caroline Rooryck-Thambo, Delphine Dupin-Deguine, et al.. FLNC pathogenic variants in patients with cardiomyopathies: Prevalence and genotype-phenotype correlations. Clinical Genetics, Wiley, 2019, 96 (4), pp.317-329. 10.1111/cge.13594. hal-02268422 HAL Id: hal-02268422 https://hal-normandie-univ.archives-ouvertes.fr/hal-02268422 Submitted on 29 Jun 2020 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. FLNC pathogenic variants in patients with cardiomyopathies Prevalence and genotype-phenotype correlations Running Title : FLNC variants genotype-phenotype correlation Flavie Ader1,2,3, Pascal De Groote4, Patricia Réant5, Caroline Rooryck-Thambo6, Delphine Dupin-Deguine7, Caroline Rambaud8, Diala Khraiche9, Claire Perret2, Jean Francois Pruny10, Michèle Mathieu Dramard11, Marion Gérard12, Yann Troadec12, Laurent Gouya13, Xavier Jeunemaitre14, Lionel Van Maldergem15, Albert Hagège16, Eric Villard2, Philippe Charron2, 10, Pascale Richard1, 2, 10. Conflict of interest statement: none declared for each author 1.
    [Show full text]
  • Genetic Variation Screening of TNNT2 Gene in a Cohort of Patients with Hypertrophic and Dilated Cardiomyopathy
    Physiol. Res. 61: 169-175, 2012 https://doi.org/10.33549/physiolres.932157 Genetic Variation Screening of TNNT2 Gene in a Cohort of Patients With Hypertrophic and Dilated Cardiomyopathy M. JÁCHYMOVÁ1, A. MURAVSKÁ1, T. PALEČEK2, P. KUCHYNKA2, H. ŘEHÁKOVÁ1, S. MAGAGE2, A. KRÁL2, T. ZIMA1, K. HORKÝ2, A. LINHART2 1Institute of Clinical Chemistry and Laboratory Diagnostics, First Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic, 2Second Department of Internal Medicine – Clinical Department of Cardiology and Angiology, First Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic Received February 1, 2011 Accepted October 17, 2011 On-line January 31, 2012 Summary Introduction Mutations in troponin T (TNNT2) gene represent the important part of currently identified disease-causing mutations in Cardiomyopathies are generally defined as hypertrophic (HCM) and dilated (DCM) cardiomyopathy. The aim myocardial disorders in which the heart muscle is of this study was to analyze TNNT2 gene exons in patients with structurally and functionally abnormal, in the absence of HCM and DCM diagnosis to improve diagnostic and genetic coronary artery disease, hypertension, valvular disease consultancy in affected families. All 15 exons and their flanking and congenital heart disease sufficient to cause the regions of the TNNT2 gene were analyzed by DNA sequence observed myocardial abnormality (Elliott et al. 2008). analysis in 174 patients with HCM and DCM diagnosis. We According to the morphological and functional phenotype identified genetic variations in TNNT2 exon regions in 56 patients the diagnosis of hypertrophic and dilated cardiomyopathy and genetic variations in TNNT2 intron regions in 164 patients.
    [Show full text]
  • Identification of the Binding Partners for Hspb2 and Cryab Reveals
    Brigham Young University BYU ScholarsArchive Theses and Dissertations 2013-12-12 Identification of the Binding arP tners for HspB2 and CryAB Reveals Myofibril and Mitochondrial Protein Interactions and Non- Redundant Roles for Small Heat Shock Proteins Kelsey Murphey Langston Brigham Young University - Provo Follow this and additional works at: https://scholarsarchive.byu.edu/etd Part of the Microbiology Commons BYU ScholarsArchive Citation Langston, Kelsey Murphey, "Identification of the Binding Partners for HspB2 and CryAB Reveals Myofibril and Mitochondrial Protein Interactions and Non-Redundant Roles for Small Heat Shock Proteins" (2013). Theses and Dissertations. 3822. https://scholarsarchive.byu.edu/etd/3822 This Thesis is brought to you for free and open access by BYU ScholarsArchive. It has been accepted for inclusion in Theses and Dissertations by an authorized administrator of BYU ScholarsArchive. For more information, please contact [email protected], [email protected]. Identification of the Binding Partners for HspB2 and CryAB Reveals Myofibril and Mitochondrial Protein Interactions and Non-Redundant Roles for Small Heat Shock Proteins Kelsey Langston A thesis submitted to the faculty of Brigham Young University in partial fulfillment of the requirements for the degree of Master of Science Julianne H. Grose, Chair William R. McCleary Brian Poole Department of Microbiology and Molecular Biology Brigham Young University December 2013 Copyright © 2013 Kelsey Langston All Rights Reserved ABSTRACT Identification of the Binding Partners for HspB2 and CryAB Reveals Myofibril and Mitochondrial Protein Interactors and Non-Redundant Roles for Small Heat Shock Proteins Kelsey Langston Department of Microbiology and Molecular Biology, BYU Master of Science Small Heat Shock Proteins (sHSP) are molecular chaperones that play protective roles in cell survival and have been shown to possess chaperone activity.
    [Show full text]
  • TNNI3 Gene Troponin I3, Cardiac Type
    TNNI3 gene troponin I3, cardiac type Normal Function The TNNI3 gene provides instructions for making a protein called cardiac troponin I, which is found solely in the heart (cardiac) muscle. Cardiac troponin I is one of three proteins that make up the troponin protein complex in cardiac muscle cells. The troponin complex is associated with a structure called the sarcomere, which is the basic unit of muscle contraction. Sarcomeres are made up of thick and thin filaments. The overlapping thick and thin filaments attach (bind) to each other and release, which allows the filaments to move relative to one another so that muscles can contract. The troponin complex, along with calcium, helps regulate tensing (contraction) of cardiac muscle. For the heart to beat normally, cardiac muscle must contract and relax in a coordinated way. Cardiac troponin I helps to coordinate contraction of the heart. When calcium levels are low, the troponin complex binds to the thin filament. This binding blocks the interaction between the thick and thin filaments that is needed for muscle contraction. An increase in calcium levels causes structural changes in another troponin complex protein called troponin C, which then triggers the troponin complex to detach from the thin filament, allowing the heart muscle to contract. Health Conditions Related to Genetic Changes Familial hypertrophic cardiomyopathy Mutations in the TNNI3 gene can cause familial hypertrophic cardiomyopathy, a condition characterized by thickening (hypertrophy) of the cardiac muscle. TNNI3 gene mutations are found in less than 5 percent of people with this condition. Although some people with hypertrophic cardiomyopathy have no obvious health effects, all affected individuals have an increased risk of heart failure and sudden death.
    [Show full text]
  • Large-Scale Serum Protein Biomarker Discovery in Duchenne Muscular Dystrophy
    Large-scale serum protein biomarker discovery in Duchenne muscular dystrophy Yetrib Hathouta, Edward Brodyb, Paula R. Clemensc,d, Linda Cripee, Robert Kirk DeLisleb, Pat Furlongf, Heather Gordish- Dressmana, Lauren Hachea, Erik Henricsong, Eric P. Hoffmana, Yvonne Monique Kobayashih, Angela Lortsi, Jean K. Mahj, Craig McDonaldg, Bob Mehlerb, Sally Nelsonk, Malti Nikradb, Britta Singerb, Fintan Steeleb, David Sterlingb, H. Lee Sweeneyl, Steve Williamsb, and Larry Goldb,1 aResearch Center for Genetic Medicine, Children’s National Medical Center, Washington, DC 20012; bSomaLogic, Inc., Boulder, CO 80301; cNeurology Service, Department of Veteran Affairs Medical Center, Pittsburgh, PA 15240; dUniversity of Pittsburgh, Pittsburgh, PA 15213; eThe Heart Center, Nationwide Children’s Hospital, The Ohio State University, Columbus, OH 15213; fParent Project Muscular Dystrophy, Hackensack, NJ 07601; gDepartment of Physical Medicine and Rehabilitation, University of California Davis School of Medicine, Davis, CA 95618; hDepartment of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202; iThe Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229; jDepartment of Pediatrics, University of Calgary, Alberta Children’s Hospital, Calgary, AB, Canada T3B 6A8; kDivision of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045; and lDepartment of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610 Contributed
    [Show full text]
  • Novel Pathogenic Variants in Filamin C Identified in Pediatric Restrictive Cardiomyopathy
    Novel pathogenic variants in filamin C identified in pediatric restrictive cardiomyopathy Jeffrey Schubert1, 2, Muhammad Tariq3, Gabrielle Geddes4, Steven Kindel4, Erin M. Miller5, and Stephanie M. Ware2. 1 Department of Molecular Genetics, Microbiology, and Biochemistry, University of Cincinnati College of Medicine, Cincinnati, OH; 2 Departments of Pediatrics and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN; 3 Faculty of Applied Medical Science, University of Tabuk, Tabuk, Kingdom of Saudi Arabia; 4Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI; 5Cincinnati Children’s Hospital Medical Center, Cincinnati, OH. Correspondence: Stephanie M. Ware, MD, PhD Department of Pediatrics Indiana University School of Medicine 1044 W. Walnut Street Indianapolis, IN 46202 Telephone: 317 274-8939 Email: [email protected] Grant Sponsor: The project was supported by the Children’s Cardiomyopathy Foundation (S.M.W.), an American Heart Association Established Investigator Award 13EIA13460001 (S.M.W.) and an AHA Postdoctoral Fellowship Award 12POST10370002 (M.T.). ___________________________________________________________________ This is the author's manuscript of the article published in final edited form as: Schubert, J., Tariq, M., Geddes, G., Kindel, S., Miller, E. M., & Ware, S. M. (2018). Novel pathogenic variants in filamin C identified in pediatric restrictive cardiomyopathy. Human Mutation, 0(ja). https://doi.org/10.1002/humu.23661 Abstract Restrictive cardiomyopathy (RCM) is a rare and distinct form of cardiomyopathy characterized by normal ventricular chamber dimensions, normal myocardial wall thickness, and preserved systolic function. The abnormal myocardium, however, demonstrates impaired relaxation. To date, dominant variants causing RCM have been reported in a small number of sarcomeric or cytoskeletal genes, but the genetic causes in a majority of cases remain unexplained especially in early childhood.
    [Show full text]
  • Troponin Variants in Congenital Myopathies: How They Affect Skeletal Muscle Mechanics
    International Journal of Molecular Sciences Review Troponin Variants in Congenital Myopathies: How They Affect Skeletal Muscle Mechanics Martijn van de Locht , Tamara C. Borsboom, Josine M. Winter and Coen A. C. Ottenheijm * Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands; [email protected] (M.v.d.L.); [email protected] (T.C.B.); [email protected] (J.M.W.) * Correspondence: [email protected]; Tel.: +31-(0)-20-444-8123 Abstract: The troponin complex is a key regulator of muscle contraction. Multiple variants in skeletal troponin encoding genes result in congenital myopathies. TNNC2 has been implicated in a novel congenital myopathy, TNNI2 and TNNT3 in distal arthrogryposis (DA), and TNNT1 and TNNT3 in nemaline myopathy (NEM). Variants in skeletal troponin encoding genes compromise sarcomere function, e.g., by altering the Ca2+ sensitivity of force or by inducing atrophy. Several potential therapeutic strategies are available to counter the effects of variants, such as troponin activators, introduction of wild-type protein through AAV gene therapy, and myosin modulation to improve muscle contraction. The mechanisms underlying the pathophysiological effects of the variants in skeletal troponin encoding genes are incompletely understood. Furthermore, limited knowledge is available on the structure of skeletal troponin. This review focusses on the physiology of slow and fast skeletal troponin and the pathophysiology of reported variants in skeletal troponin encoding genes. A better understanding of the pathophysiological effects of these variants, together with enhanced knowledge regarding the structure of slow and fast skeletal troponin, will direct the development of Citation: van de Locht, M.; treatment strategies.
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • Bovine Cardiac Troponin I Gene (TNNI3) As a Candidate Gene for Bovine Dilated Cardiomyopathy
    University of Pennsylvania ScholarlyCommons Departmental Papers (Vet) School of Veterinary Medicine 2009 Bovine Cardiac Troponin I Gene (TNNI3) as a Candidate Gene for Bovine Dilated Cardiomyopathy Marta Owczarek-Lipska Gaudenz Dolf Karina E. Guziewicz University of Pennsylvania Tosso Leeb Claude Shelling See next page for additional authors Follow this and additional works at: https://repository.upenn.edu/vet_papers Part of the Large or Food Animal and Equine Medicine Commons Recommended Citation Owczarek-Lipska, M., Dolf, G., Guziewicz, K. E., Leeb, T., Shelling, C., Posthaus, H., & Braunschweig, M. H. (2009). Bovine Cardiac Troponin I Gene (TNNI3) as a Candidate Gene for Bovine Dilated Cardiomyopathy. Archiv Tierzucht (Archives Animal Breeding), 52 (2), 113-123. Retrieved from https://repository.upenn.edu/vet_papers/28 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/vet_papers/28 For more information, please contact [email protected]. Bovine Cardiac Troponin I Gene (TNNI3) as a Candidate Gene for Bovine Dilated Cardiomyopathy Abstract The cardiac troponin complex, which is an important component of the contractile apparatus, is composed of the three subunits troponin I (TnI), troponin C (TnC) and troponin T (TnT). Troponin I is the inhibitory subunit and consists of three isoforms encoded by TNNI1, TNNI2 and TNNI3 genes, respectively. Due to the different types of cardiomyopathies caused by mutations in the TNNI3 gene and its fluorescence in situ hybridization (FISH) mapping on bovine chromosome 18q26, which was shown to be linked to the recessively inherited bovine dilated cardiomyopathy (BDCMP), bovine TNNI3 was considered as candidate gene for BDCMP. Real-time polymerase chain reaction (PCR) TNNI3 expression analysis resulted in a significant difference between BDCMP affected and unaffected animals when normalized to ACTB gene expression, but there was no significant difference in expression when normalized to GAPDH.
    [Show full text]
  • Are These Cardiomyocytes? Protocol Development Reveals Impact of Sample Preparation on the Accuracy of Identifying Cardiomyocytes by Flow Cytometry
    bioRxiv preprint doi: https://doi.org/10.1101/388926; this version posted August 9, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Resource Are these cardiomyocytes? Protocol development reveals impact of sample preparation on the accuracy of identifying cardiomyocytes by flow cytometry Matthew Waas1, Ranjuna Weerasekera1, Erin M. Kropp1, Marisol Romero-Tejeda2,3, Ellen Poon4, Kenneth R. Boheler4,5,6, Paul W. Burridge2,3, Rebekah L. Gundry1,7 1Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA 2Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA 3Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA 4School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China. 5Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. 6Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA. 7Center for Biomedical Mass Spectrometry Research, Medical College of Wisconsin, Milwaukee, WI 53226, USA Corresponding author: Rebekah L. Gundry, PhD Department of Biochemistry Medical College of Wisconsin 8701 Watertown Plank Road Milwaukee, WI, 53226 Telephone: 414-955-2825 Fax: 414-955-6568 Email: [email protected] Running Title: Standard flow cytometry protocol for assessing stem cell-derived cardiomyocytes bioRxiv preprint doi: https://doi.org/10.1101/388926; this version posted August 9, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • Chamber-Enriched Gene Expression Profiles in Failing Human Hearts with Reduced Ejection Fraction
    www.nature.com/scientificreports OPEN Chamber‑enriched gene expression profles in failing human hearts with reduced ejection fraction Xin Luo1, Jun Yin1, Denise Dwyer2, Tracy Yamawaki1, Hong Zhou1, Hongfei Ge3, Chun‑Ya Han2, Artem Shkumatov4, Karen Snyder5, Brandon Ason3, Chi‑Ming Li1, Oliver Homann1 & Marina Stolina2* Heart failure with reduced ejection fraction (HFrEF) constitutes 50% of HF hospitalizations and is characterized by high rates of mortality. To explore the underlying mechanisms of HFrEF etiology and progression, we studied the molecular and cellular diferences in four chambers of non‑failing (NF, n = 10) and HFrEF (n = 12) human hearts. We identifed 333 genes enriched within NF heart subregions and often associated with cardiovascular disease GWAS variants. Expression analysis of HFrEF tissues revealed extensive disease‑associated transcriptional and signaling alterations in left atrium (LA) and left ventricle (LV). Common left heart HFrEF pathologies included mitochondrial dysfunction, cardiac hypertrophy and fbrosis. Oxidative stress and cardiac necrosis pathways were prominent within LV, whereas TGF‑beta signaling was evident within LA. Cell type composition was estimated by deconvolution and revealed that HFrEF samples had smaller percentage of cardiomyocytes within the left heart, higher representation of fbroblasts within LA and perivascular cells within the left heart relative to NF samples. We identifed essential modules associated with HFrEF pathology and linked transcriptome discoveries with human genetics fndings. This study contributes to a growing body of knowledge describing chamber‑specifc transcriptomics and revealed genes and pathways that are associated with heart failure pathophysiology, which may aid in therapeutic target discovery. By physiological function, the human heart is a muscular pump that circulates blood, perfusing tissues through- out the body.
    [Show full text]
  • Elevation of Fast but Not Slow Troponin I in the Circulation of Patients With
    Russell Alan (Orcid ID: 0000-0001-7545-5008) Elevation of fast but not slow troponin I in the circulation of patients with Becker and Duchenne muscular dystrophy Benjamin L Barthel PhD1, Dan Cox BSc2, Marissa Barbieri BA3, Michael Ziemba3, Volker Straub MD, PhD2, Eric P. Hoffman PhD3, Alan J Russell PhD1 1Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA. 2The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE1 3BZ, UK 3Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University – State University of New York, Binghamton, NY 13902 USA Acknowledgements: Source of funding, private industry Number of words in abstract: 220 Number of words in manuscript: 2490 Corresponding author: Alan J Russell, 1Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA. [email protected]. ORCID ID https://orcid.org/0000-0001-7545-5008 Ethical publication statement: We confirm that we have read the Journal’s position on issues involved in ethical publication and affirm that this report is consistent This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process which may lead to differences between this version and the Version of Record. Please cite this article as doi: 10.1002/mus.27222 This article is protected by copyright. All rights reserved. with those guidelines. Key words: Muscular dystrophy, biomarker, muscle injury, creatine kinase, troponin Conflicts of interest: Benjamin Barthel and Alan Russell are paid employees of Edgewise Therapeutics, Inc.
    [Show full text]