1
|
Khan B, Lanzuolo C, Rosti V, Santarelli P, Pich A, Kraft T, Amrute-Nayak M, Nayak A. Sorafenib induces cachexia by impeding transcriptional signaling of the SET1/MLL complex on muscle-specific genes. iScience 2024; 27:110913. [PMID: 39386761 PMCID: PMC11462028 DOI: 10.1016/j.isci.2024.110913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/27/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
Chemotherapeutics used in cancer therapy are often linked to muscle wasting or cachexia. Insights into the molecular basis of chemotherapy-induced cachexia is essential to improve treatment strategies. Here, we demonstrated that Sorafenib-tyrosine kinase inhibitor (TKI) class of chemotherapeutic agents-induced cachexia. System-wide analyses revealed that Sorafenib alters the global transcriptional program and proteostasis in muscle cells. Mechanistically, Sorafenib treatment reduced active epigenetic mark H3K4 methylation on distinct muscle-specific genes by impeding chromatin association of SET1A-catalytic component of the SET1/MLL histone methyltransferase complex. This mechanism favored transcriptional disorientation that led to disrupted sarcomere assembly, calcium homeostasis and mitochondrial respiration. Consequently, the contractile ability of muscle cells was severely compromised. Interestingly, the other prominent TKIs Nilotinib and Imatinib did not exert similar effects on muscle cell physiology. Collectively, we identified an unanticipated transcriptional mechanism underlying Sorafenib-induced cachexia. Our findings hold the potential to strategize therapy regimens to minimize chemotherapy-induced cachexia.
Collapse
Affiliation(s)
- Bushra Khan
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Chiara Lanzuolo
- Istituto Nazionale Genetica Molecolare ‘Romeo ed Enrica Invernizzi’, Milan, Italy
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Valentina Rosti
- Istituto Nazionale Genetica Molecolare ‘Romeo ed Enrica Invernizzi’, Milan, Italy
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Philina Santarelli
- Istituto Nazionale Genetica Molecolare ‘Romeo ed Enrica Invernizzi’, Milan, Italy
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Andreas Pich
- Institute of Toxicology, Core Facility Proteomics, Hannover Medical School, Hannover, Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Mamta Amrute-Nayak
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Arnab Nayak
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
2
|
Htet M, Lei S, Bajpayi S, Gangrade H, Arvanitis M, Zoitou A, Murphy S, Chen EZ, Koleini N, Lin BL, Kwon C, Tampakakis E. A transcriptional enhancer regulates cardiac maturation. NATURE CARDIOVASCULAR RESEARCH 2024; 3:666-684. [PMID: 39196225 DOI: 10.1038/s44161-024-00484-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 05/02/2024] [Indexed: 08/29/2024]
Abstract
Cardiomyocyte maturation is crucial for generating adult cardiomyocytes and the application of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs). However, regulation at the cis-regulatory element level and its role in heart disease remain unclear. Alpha-actinin 2 (ACTN2) levels increase during CM maturation. In this study, we investigated a clinically relevant, conserved ACTN2 enhancer's effects on CM maturation using hPSC and mouse models. Heterozygous ACTN2 enhancer deletion led to abnormal CM morphology, reduced function and mitochondrial respiration. Transcriptomic analyses in vitro and in vivo showed disrupted CM maturation and upregulated anabolic mammalian target for rapamycin (mTOR) signaling, promoting senescence and hindering maturation. As confirmation, ACTN2 enhancer deletion induced heat shock protein 90A expression, a chaperone mediating mTOR activation. Conversely, targeting the ACTN2 enhancer via enhancer CRISPR activation (enCRISPRa) promoted hPSC-CM maturation. Our studies reveal the transcriptional enhancer's role in cardiac maturation and disease, offering insights into potentially fine-tuning gene expression to modulate cardiomyocyte physiology.
Collapse
Grants
- K99 HL155840 NHLBI NIH HHS
- 2023- MSCRFL-5984 Maryland Stem Cell Research Fund (MSCRF)
- 5K08HL166690 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- CDA34660077 American Heart Association (American Heart Association, Inc.)
- R00 HL155840 NHLBI NIH HHS
- TPA1058685 American Heart Association (American Heart Association, Inc.)
- T32HL007227 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL-145135 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL156947 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K08 HL145135 NHLBI NIH HHS
- MSCRFD-6139 Maryland Stem Cell Research Fund (MSCRF)
Collapse
Affiliation(s)
- Myo Htet
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Shunyao Lei
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sheetal Bajpayi
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Harshi Gangrade
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Marios Arvanitis
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Asimina Zoitou
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sean Murphy
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Elaine Zhelan Chen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Navid Koleini
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Brian Leei Lin
- Department of Cell Biology, Neurobiology, and Anatomy and Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Chulan Kwon
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute of Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Emmanouil Tampakakis
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
3
|
Jones TLM, Woulfe KC. Considering impact of age and sex on cardiac cytoskeletal components. Am J Physiol Heart Circ Physiol 2024; 326:H470-H478. [PMID: 38133622 PMCID: PMC11219061 DOI: 10.1152/ajpheart.00619.2023] [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: 10/02/2023] [Revised: 12/08/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023]
Abstract
The cardiac cytoskeletal components are integral to cardiomyocyte function and are responsible for contraction, sustaining cell structure, and providing scaffolding to direct signaling. Cytoskeletal components have been implicated in cardiac pathology; however, less attention has been paid to age-related modifications of cardiac cytoskeletal components and how these contribute to dysfunction with increased age. Moreover, significant sex differences in cardiac aging have been identified, but we still lack a complete understanding to the mechanisms behind these differences. This review summarizes what is known about how key cardiomyocyte cytoskeletal components are modified because of age, as well as reported sex-specific differences. Thorough consideration of both age and sex as integral players in cytoskeletal function may reveal potential avenues for more personalized therapeutics.
Collapse
Affiliation(s)
- Timothy L M Jones
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kathleen C Woulfe
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
4
|
Qi T, Zhang J, Zhang K, Zhang W, Song Y, Lian K, Kan C, Han F, Hou N, Sun X. Unraveling the role of the FHL family in cardiac diseases: Mechanisms, implications, and future directions. Biochem Biophys Res Commun 2024; 694:149468. [PMID: 38183876 DOI: 10.1016/j.bbrc.2024.149468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
Heart diseases are a major cause of morbidity and mortality worldwide. Understanding the molecular mechanisms underlying these diseases is essential for the development of effective diagnostic and therapeutic strategies. The FHL family consists of five members: FHL1, FHL2, FHL3, FHL4, and FHL5/Act. These members exhibit different expression patterns in various tissues including the heart. FHL family proteins are implicated in cardiac remodeling, regulation of metabolic enzymes, and cardiac biomechanical stress perception. A large number of studies have explored the link between FHL family proteins and cardiac disease, skeletal muscle disease, and ovarian metabolism, but a comprehensive and in-depth understanding of the specific molecular mechanisms targeting FHL on cardiac disease is lacking. The aim of this review is to explore the structure and function of FHL family members, to comprehensively elucidate the mechanisms by which they regulate the heart, and to explore in depth the changes in FHL family members observed in different cardiac disorders, as well as the effects of mutations in FHL proteins on heart health.
Collapse
Affiliation(s)
- Tongbing Qi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Wenqiang Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Yixin Song
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Kexin Lian
- Department of Nephrology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| |
Collapse
|
5
|
Vasileva F, Font-Lladó R, Carreras-Badosa G, Roman-Viñas B, Cadellans-Arróniz A, López-Bermejo A, Prats-Puig A. Salivary cardiac-enriched FHL2-interacting protein is associated with higher diastolic-to-systolic-blood pressure ratio, sedentary time and center of pressure displacement in healthy 7-9 years old school-children. Front Endocrinol (Lausanne) 2024; 15:1292653. [PMID: 38304464 PMCID: PMC10830845 DOI: 10.3389/fendo.2024.1292653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/04/2024] [Indexed: 02/03/2024] Open
Abstract
Introduction Cardiac-enriched FHL2-interacting protein (CEFIP) is a recently identified protein, first found in the z-disc of striated muscles, and related to cardiovascular diseases. Our objectives are: 1) to quantify CEFIP in saliva in healthy 7-9 years old school-children; and 2) to assess the associations of salivary CEFIP concentration and blood pressure, physical (in)activity and physical fitness in these children. Methods A total of 72 children (7.6 ± 0.3 years) were included in the study, recruited in primary schools in Girona (Spain). A sandwich enzyme-linked immunosorbent assay was used (abx506878; Abbexa, United Kingdom) to quantify CEFIP in saliva. Anthropometric evaluation was performed [body mass, height and body mass index (BMI)]. Systolic and diastolic blood pressure were measured by means of an electronic oscillometer and the diastolic-to-systolic blood pressure ratio (D/S BP ratio) was calculated. Physical (in)activity [sedentary time and time spent in physical activity (PA)] were assessed by means of a triaxial Actigraph GT3X accelerometer (Actigraph, Pensacola, FL, USA) that children were instructed to wear for 24h during 7 conssecutive days. Finally, physical fitness (speed and agility, explosive power of legs, handgrip strength, flexibility and balance) were assessed through validated and standardized testing batteries. Results CEFIP was easily detected and measured in all saliva samples (mean concentration: 0.6 ± 0.2 pg/ml). Salivary CEFIP was positively associated with D/S BP ratio (r=0.305, p=0.010) and sedentary time (r=0.317, p=0.012), but negatively associated with PA in 7-9 years old school-children (r=-0.350, p=0.002). Furthermore, salivary CEFIP was related to lower level of balance i.e., higher center of pressure (CoP) displacement in these children (r=0.411, p<0.001). The associations of salivary CEFIP with D/S BP ratio (Beta=0.349, p=0.004), sedentary time (Beta=0.354, p=0.009) and CoP displacement (Beta=0.401, p=0.001), were maintained significant after adjustment for potential confounding variables such as age, gender and BMI in linear regression analyses. Conclusion CEFIP can be easily assessed in saliva as a promising biomarker associated with cardiovascular health in 7-9 years old school-children. Interestingly, higher salivary CEFIP concentration was related to higher D/S BP ratio, more sedentary time and higher CoP displacement i.e., lower level of balance in these children.
Collapse
Affiliation(s)
- Fidanka Vasileva
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research, Girona, Spain
- University School of Health and Sport, University of Girona, Girona, Spain
| | - Raquel Font-Lladó
- University School of Health and Sport, University of Girona, Girona, Spain
- Research Group of Culture and Education, Institute of Educational Research, University of Girona, Girona, Spain
| | - Gemma Carreras-Badosa
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research, Girona, Spain
| | - Blanca Roman-Viñas
- University School of Health and Sport, University of Girona, Girona, Spain
- Department of Physical Activity and Sport Sciences, Blanquerna-Universitat Ramon Llull, Barcelona, Spain
| | - Aïda Cadellans-Arróniz
- Department of Physiotherapy, Faculty of Medicine and Health Sciences, International University of Catalonia, Barcelona, Spain
| | - Abel López-Bermejo
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research, Girona, Spain
- Department of Medical Sciences, University of Girona, Girona, Spain
- Pediatric Endocrinology, Dr. Josep Trueta Hospital, Girona, Spain
| | - Anna Prats-Puig
- University School of Health and Sport, University of Girona, Girona, Spain
- Research Group of Clinical Anatomy, Embryology and Neuroscience, Department of Medical Sciences, University of Girona, Girona, Spain
| |
Collapse
|
6
|
Ormrod B, Ehler E. Induced pluripotent stem cell-derived cardiomyocytes-more show than substance? Biophys Rev 2023; 15:1941-1950. [PMID: 38192353 PMCID: PMC10771368 DOI: 10.1007/s12551-023-01099-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/04/2023] [Indexed: 01/10/2024] Open
Abstract
Cardiomyocytes that are derived from human-induced pluripotent stem cells (iPSC-CM) are an exciting tool to investigate cardiomyopathy disease mechanisms at the cellular level as well as to screen for potential side effects of novel drugs. However, currently their benefit is limited due to their fairly immature differentiation status under conventional culture conditions. This review is mainly aimed at researchers outside of the iPSC-CM field and will describe potential pitfalls and which features at the level of the myofibrils would be desired to make them a more representative model system. We will also discuss different strategies that may help to achieve these.
Collapse
Affiliation(s)
- Beth Ormrod
- School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, SE1 1UL UK
| | - Elisabeth Ehler
- School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, SE1 1UL UK
- Randall Centre for Cell and Molecular Biophysics (School of Basic and Biosciences), Room 3.26A, New Hunt’s House, Guy’s Campus, London, SE1 1UL UK
- British Heart Foundation Centre of Research Excellence, King’s College London, London, SE1 1UL UK
| |
Collapse
|
7
|
Yeganeh FA, Summerill C, Hu Z, Rahmani H, Taylor DW, Taylor KA. The cryo-EM 3D image reconstruction of isolated Lethocerus indicus Z-discs. J Muscle Res Cell Motil 2023; 44:271-286. [PMID: 37661214 PMCID: PMC10843718 DOI: 10.1007/s10974-023-09657-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 08/14/2023] [Indexed: 09/05/2023]
Abstract
The Z-disk of striated muscle defines the ends of the sarcomere, which repeats many times within the muscle fiber. Here we report application of cryoelectron tomography and subtomogram averaging to Z-disks isolated from the flight muscles of the large waterbug Lethocerus indicus. We use high salt solutions to remove the myosin containing filaments and use gelsolin to remove the actin filaments of the A- and I-bands leaving only the thin filaments within the Z-disk which were then frozen for cryoelectron microscopy. The Lethocerus Z-disk structure is similar in many ways to the previously studied Z-disk of the honeybee Apis mellifera. At the corners of the unit cell are positioned trimers of paired antiparallel F-actins defining a large solvent channel, whereas at the trigonal positions are positioned F-actin trimers converging slowly towards their (+) ends defining a small solvent channel through the Z-disk. These near parallel F-actins terminate at different Z-heights within the Z-disk. The two types of solvent channel in Lethocerus are similar in size compared to those of Apis which are very different in size. Two types of α-actinin crosslinks were observed between oppositely oriented actin filaments. In one of these, the α-actinin long axis is almost parallel to the F-actins it crosslinks. In the other, the α-actinins are at a small but distinctive angle with respect to the crosslinked actin filaments. The utility of isolated Z-disks for structure determination is discussed.
Collapse
Affiliation(s)
- Fatemeh Abbasi Yeganeh
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32306-4380, USA
| | - Corinne Summerill
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32306-4380, USA
- Department of Life and Earth Sciences, Perimeter College, Georgia State University, 33 Gilmer Street SE, Atlanta, GA, 30303, USA
| | - Zhongjun Hu
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32306-4380, USA
- Facebook, Inc, 1 Hacker Way, Menlo Park, CA, 94025, USA
| | - Hamidreza Rahmani
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32306-4380, USA
- The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Dianne W Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32306-4380, USA
| | - Kenneth A Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32306-4380, USA.
| |
Collapse
|
8
|
Wang S, Zhang Z, He J, Liu J, Guo X, Chu H, Xu H, Wang Y. Comprehensive review on gene mutations contributing to dilated cardiomyopathy. Front Cardiovasc Med 2023; 10:1296389. [PMID: 38107262 PMCID: PMC10722203 DOI: 10.3389/fcvm.2023.1296389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the most common primary myocardial diseases. However, to this day, it remains an enigmatic cardiovascular disease (CVD) characterized by ventricular dilatation, which leads to myocardial contractile dysfunction. It is the most common cause of chronic congestive heart failure and the most frequent indication for heart transplantation in young individuals. Genetics and various other factors play significant roles in the progression of dilated cardiomyopathy, and variants in more than 50 genes have been associated with the disease. However, the etiology of a large number of cases remains elusive. Numerous studies have been conducted on the genetic causes of dilated cardiomyopathy. These genetic studies suggest that mutations in genes for fibronectin, cytoskeletal proteins, and myosin in cardiomyocytes play a key role in the development of DCM. In this review, we provide a comprehensive description of the genetic basis, mechanisms, and research advances in genes that have been strongly associated with DCM based on evidence-based medicine. We also emphasize the important role of gene sequencing in therapy for potential early diagnosis and improved clinical management of DCM.
Collapse
Affiliation(s)
- Shipeng Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zhiyu Zhang
- Department of Cardiovascular Medicine, The Second People's Hospital of Yibin, Yibin, China
| | - Jiahuan He
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Junqian Liu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xia Guo
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Haoxuan Chu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Hanchi Xu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yushi Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
9
|
Zhang L, Qi D, Peng M, Meng B, Wang X, Zhang X, Zuo Z, Li L, Wang Z, Zou W, Hu Z, Qian Z. Decoding molecular signature on heart of septic mice with distinct left ventricular ejection fraction. iScience 2023; 26:107825. [PMID: 37736036 PMCID: PMC10509301 DOI: 10.1016/j.isci.2023.107825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/07/2023] [Accepted: 08/31/2023] [Indexed: 09/23/2023] Open
Abstract
Dysregulated cardiac function after sepsis in intensive care unit is known to predict poor long-term outcome and increase mortality. Their pathological feature and molecular mechanism remain unclear. We observed that septic patients with depressed left ventricular ejection fraction (LVEF) have the highest in-hospital and 28 days mortality comparing to patients with hyperdynamic LVEF or with heart failure with preserved LVEF. Echocardiograms reveal that survivors post cecum ligation and puncture (CLP) on rodents have stable LVEF and non-survivors have fluctuated LVEF at CLP early phase. CLP-induced mice fall into three groups based on LVEF 24 h post-surgery: high-, low-, and normal-LVEF. Transcriptomic and proteomic analyses identify jointly and distinctively changed genes, proteins and biologically essential pathways in left ventricles from three CLP groups. Notably, transmission electron microscopy shows different mitochondrial and sarcomere defects associated with LVEF variances. Together, this study systematically characterizes the molecular, morphological, and functional alterations in CLP-induced cardiac injury.
Collapse
Affiliation(s)
- Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Desheng Qi
- Department of Emergency Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Milin Peng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Binbin Meng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xinrun Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiaolei Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhihong Zuo
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Li Li
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Zhanwen Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Wenxuan Zou
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zhonghua Hu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhaoxin Qian
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| |
Collapse
|
10
|
Petersen M, Schmiedel N, Dierck F, Hille S, Remes A, Senger F, Schmidt I, Lüllmann-Rauch R, Müller OJ, Frank D, Rangrez AY, Frey N, Kuhn C. Fibin regulates cardiomyocyte hypertrophy and causes protein-aggregate-associated cardiomyopathy in vivo. Front Mol Biosci 2023; 10:1169658. [PMID: 37342207 PMCID: PMC10278231 DOI: 10.3389/fmolb.2023.1169658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/16/2023] [Indexed: 06/22/2023] Open
Abstract
Despite the identification of numerous molecular pathways modulating cardiac hypertrophy its pathogenesis is not completely understood. In this study we define an unexpected role for Fibin ("fin bud initiation factor homolog") in cardiomyocyte hypertrophy. Via gene expression profiling in hypertrophic murine hearts after transverse aortic constriction we found a significant induction of Fibin. Moreover, Fibin was upregulated in another mouse model of cardiac hypertrophy (calcineurin-transgenics) as well as in patients with dilated cardiomyopathy. Immunoflourescence microscopy revealed subcellular localization of Fibin at the sarcomeric z-disc. Overexpression of Fibin in neonatal rat ventricular cardiomyocytes revealed a strong anti-hypertrophic effect through inhibiting both, NFAT- and SRF-dependent signalling. In contrast, transgenic mice with cardiac-restricted overexpression of Fibin developed dilated cardiomyopathy, accompanied by induction of hypertrophy-associated genes. Moreover, Fibin overexpression accelerated the progression to heart failure in the presence of prohypertrophic stimuli such as pressure overload and calcineurin overexpression. Histological and ultrastructural analyses surprisingly showed large protein aggregates containing Fibin. On the molecular level, aggregate formation was accompanied by an induction of the unfolded protein response subsequent UPR-mediated apoptosis and autophagy. Taken together, we identified Fibin as a novel potent negative regulator of cardiomyocyte hypertrophy in vitro. Yet, heart-specific Fibin overexpression in vivo causes development of a protein-aggregate-associated cardiomyopathy. Because of close similarities to myofibrillar myopathies, Fibin represents a candidate gene for cardiomyopathy and Fibin transgenic mice may provide additional mechanistic insight into aggregate formation in these diseases.
Collapse
Affiliation(s)
- Matthias Petersen
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Nesrin Schmiedel
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Franziska Dierck
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Susanne Hille
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Anca Remes
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Frauke Senger
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Inga Schmidt
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | | | - Oliver J. Müller
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Derk Frank
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Ashraf Y. Rangrez
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Christian Kuhn
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
11
|
Sellung D, Heil L, Daya N, Jacobsen F, Mertens-Rill J, Zhuge H, Döring K, Piran M, Milting H, Unger A, Linke WA, Kley R, Preusse C, Roos A, Fürst DO, Ven PFMVD, Vorgerd M. Novel Filamin C Myofibrillar Myopathy Variants Cause Different Pathomechanisms and Alterations in Protein Quality Systems. Cells 2023; 12:cells12091321. [PMID: 37174721 PMCID: PMC10177260 DOI: 10.3390/cells12091321] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Myofibrillar myopathies (MFM) are a group of chronic muscle diseases pathophysiologically characterized by accumulation of protein aggregates and structural failure of muscle fibers. A subtype of MFM is caused by heterozygous mutations in the filamin C (FLNC) gene, exhibiting progressive muscle weakness, muscle structural alterations and intracellular protein accumulations. Here, we characterize in depth the pathogenicity of two novel truncating FLNc variants (p.Q1662X and p.Y2704X) and assess their distinct effect on FLNc stability and distribution as well as their impact on protein quality system (PQS) pathways. Both variants cause a slowly progressive myopathy with disease onset in adulthood, chronic myopathic alterations in muscle biopsy including the presence of intracellular protein aggregates. Our analyses revealed that p.Q1662X results in FLNc haploinsufficiency and p.Y2704X in a dominant-negative FLNc accumulation. Moreover, both protein-truncating variants cause different PQS alterations: p.Q1662X leads to an increase in expression of several genes involved in the ubiquitin-proteasome system (UPS) and the chaperone-assisted selective autophagy (CASA) system, whereas p.Y2704X results in increased abundance of proteins involved in UPS activation and autophagic buildup. We conclude that truncating FLNC variants might have different pathogenetic consequences and impair PQS function by diverse mechanisms and to varying extents. Further studies on a larger number of patients are necessary to confirm our observations.
Collapse
Affiliation(s)
- Dominik Sellung
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Lorena Heil
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Nassam Daya
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Frank Jacobsen
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Janine Mertens-Rill
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Heidi Zhuge
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Kristina Döring
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Misagh Piran
- Erich and Hanna Klessmann Institute, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Andreas Unger
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Wolfgang A Linke
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Rudi Kley
- Department of Neurology and Clinical Neurophysiology, St. Marien-Hospital Borken, 46325 Borken, Germany
| | - Corinna Preusse
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Andreas Roos
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| |
Collapse
|
12
|
Dowling P, Gargan S, Swandulla D, Ohlendieck K. Fiber-Type Shifting in Sarcopenia of Old Age: Proteomic Profiling of the Contractile Apparatus of Skeletal Muscles. Int J Mol Sci 2023; 24:2415. [PMID: 36768735 PMCID: PMC9916839 DOI: 10.3390/ijms24032415] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
The progressive loss of skeletal muscle mass and concomitant reduction in contractile strength plays a central role in frailty syndrome. Age-related neuronal impairments are closely associated with sarcopenia in the elderly, which is characterized by severe muscular atrophy that can considerably lessen the overall quality of life at old age. Mass-spectrometry-based proteomic surveys of senescent human skeletal muscles, as well as animal models of sarcopenia, have decisively improved our understanding of the molecular and cellular consequences of muscular atrophy and associated fiber-type shifting during aging. This review outlines the mass spectrometric identification of proteome-wide changes in atrophying skeletal muscles, with a focus on contractile proteins as potential markers of changes in fiber-type distribution patterns. The observed trend of fast-to-slow transitions in individual human skeletal muscles during the aging process is most likely linked to a preferential susceptibility of fast-twitching muscle fibers to muscular atrophy. Studies with senescent animal models, including mostly aged rodent skeletal muscles, have confirmed fiber-type shifting. The proteomic analysis of fast versus slow isoforms of key contractile proteins, such as myosin heavy chains, myosin light chains, actins, troponins and tropomyosins, suggests them as suitable bioanalytical tools of fiber-type transitions during aging.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Dieter Swandulla
- Institute of Physiology, University of Bonn, D53115 Bonn, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| |
Collapse
|
13
|
Hinkle ER, Blue RE, Tsai YH, Combs M, Davi J, Coffey AR, Boriek AM, Taylor JM, Parker JS, Giudice J. Stretching muscle cells induces transcriptional and splicing transitions and changes in SR proteins. Commun Biol 2022; 5:987. [PMID: 36123433 PMCID: PMC9485123 DOI: 10.1038/s42003-022-03915-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/30/2022] [Indexed: 11/08/2022] Open
Abstract
Alternative splicing is an RNA processing mechanism involved in skeletal muscle development and pathology. Muscular diseases exhibit splicing alterations and changes in mechanobiology leading us to investigate the interconnection between mechanical forces and RNA processing. We performed deep RNA-sequencing after stretching muscle cells. First, we uncovered transcriptional changes in genes encoding proteins involved in muscle function and transcription. Second, we observed that numerous mechanosensitive genes were part of the MAPK pathway which was activated in response to stretching. Third, we revealed that stretching skeletal muscle cells increased the proportion of alternatively spliced cassette exons and their inclusion. Fourth, we demonstrated that the serine and arginine-rich proteins exhibited stronger transcriptional changes than other RNA-binding proteins and that SRSF4 phosphorylation is mechanosensitive. Identifying SRSF4 as a mechanosensitive RNA-binding protein that might contribute to crosstalk between mechanotransduction, transcription, and splicing could potentially reveal novel insights into muscular diseases, particularly those with unknown etiologies.
Collapse
Affiliation(s)
- Emma R Hinkle
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
| | - R Eric Blue
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
| | - Yi-Hsuan Tsai
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
| | - Matthew Combs
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
| | - Jacquelyn Davi
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
| | - Alisha R Coffey
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
| | - Aladin M Boriek
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Joan M Taylor
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
| | - Joel S Parker
- Curriculum in Genetics and Molecular Biology (GMB), The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
| | - Jimena Giudice
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA.
- Curriculum in Genetics and Molecular Biology (GMB), The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA.
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA.
| |
Collapse
|
14
|
Suay-Corredera C, Alegre-Cebollada J. The mechanics of the heart: zooming in on hypertrophic cardiomyopathy and cMyBP-C. FEBS Lett 2022; 596:703-746. [PMID: 35224729 DOI: 10.1002/1873-3468.14301] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 11/10/2022]
Abstract
Hypertrophic cardiomyopathy (HCM), a disease characterized by cardiac muscle hypertrophy and hypercontractility, is the most frequently inherited disorder of the heart. HCM is mainly caused by variants in genes encoding proteins of the sarcomere, the basic contractile unit of cardiomyocytes. The most frequently mutated among them is MYBPC3, which encodes cardiac myosin-binding protein C (cMyBP-C), a key regulator of sarcomere contraction. In this review, we summarize clinical and genetic aspects of HCM and provide updated information on the function of the healthy and HCM sarcomere, as well as on emerging therapeutic options targeting sarcomere mechanical activity. Building on what is known about cMyBP-C activity, we examine different pathogenicity drivers by which MYBPC3 variants can cause disease, focussing on protein haploinsufficiency as a common pathomechanism also in nontruncating variants. Finally, we discuss recent evidence correlating altered cMyBP-C mechanical properties with HCM development.
Collapse
|
15
|
Pozo MR, Meredith GW, Entcheva E. Human iPSC-Cardiomyocytes as an Experimental Model to Study Epigenetic Modifiers of Electrophysiology. Cells 2022; 11:200. [PMID: 35053315 PMCID: PMC8774228 DOI: 10.3390/cells11020200] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/31/2021] [Accepted: 01/01/2022] [Indexed: 02/04/2023] Open
Abstract
The epigenetic landscape and the responses to pharmacological epigenetic regulators in each human are unique. Classes of epigenetic writers and erasers, such as histone acetyltransferases, HATs, and histone deacetylases, HDACs, control DNA acetylation/deacetylation and chromatin accessibility, thus exerting transcriptional control in a tissue- and person-specific manner. Rapid development of novel pharmacological agents in clinical testing-HDAC inhibitors (HDACi)-targets these master regulators as common means of therapeutic intervention in cancer and immune diseases. The action of these epigenetic modulators is much less explored for cardiac tissue, yet all new drugs need to be tested for cardiotoxicity. To advance our understanding of chromatin regulation in the heart, and specifically how modulation of DNA acetylation state may affect functional electrophysiological responses, human-induced pluripotent stem-cell-derived cardiomyocyte (hiPSC-CM) technology can be leveraged as a scalable, high-throughput platform with ability to provide patient-specific insights. This review covers relevant background on the known roles of HATs and HDACs in the heart, the current state of HDACi development, applications, and any adverse cardiac events; it also summarizes relevant differential gene expression data for the adult human heart vs. hiPSC-CMs along with initial transcriptional and functional results from using this new experimental platform to yield insights on epigenetic control of the heart. We focus on the multitude of methodologies and workflows needed to quantify responses to HDACis in hiPSC-CMs. This overview can help highlight the power and the limitations of hiPSC-CMs as a scalable experimental model in capturing epigenetic responses relevant to the human heart.
Collapse
Affiliation(s)
| | | | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC 20052, USA; (M.R.P.); (G.W.M.)
| |
Collapse
|
16
|
Solís C, Russell B. Striated muscle proteins are regulated both by mechanical deformation and by chemical post-translational modification. Biophys Rev 2021; 13:679-695. [PMID: 34777614 PMCID: PMC8555064 DOI: 10.1007/s12551-021-00835-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/23/2021] [Indexed: 01/09/2023] Open
Abstract
All cells sense force and build their cytoskeleton to optimize function. How is this achieved? Two major systems are involved. The first is that load deforms specific protein structures in a proportional and orientation-dependent manner. The second is post-translational modification of proteins as a consequence of signaling pathway activation. These two processes work together in a complex way so that local subcellular assembly as well as overall cell function are controlled. This review discusses many cell types but focuses on striated muscle. Detailed information is provided on how load deforms the structure of proteins in the focal adhesions and filaments, using α-actinin, vinculin, talin, focal adhesion kinase, LIM domain-containing proteins, filamin, myosin, titin, and telethonin as examples. Second messenger signals arising from external triggers are distributed throughout the cell causing post-translational or chemical modifications of protein structures, with the actin capping protein CapZ and troponin as examples. There are numerous unanswered questions of how mechanical and chemical signals are integrated by muscle proteins to regulate sarcomere structure and function yet to be studied. Therefore, more research is needed to see how external triggers are integrated with local tension generated within the cell. Nonetheless, maintenance of tension in the sarcomere is the essential and dominant mechanism, leading to the well-known phrase in exercise physiology: "use it or lose it."
Collapse
Affiliation(s)
- Christopher Solís
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Brenda Russell
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| |
Collapse
|
17
|
Höhfeld J, Benzing T, Bloch W, Fürst DO, Gehlert S, Hesse M, Hoffmann B, Hoppe T, Huesgen PF, Köhn M, Kolanus W, Merkel R, Niessen CM, Pokrzywa W, Rinschen MM, Wachten D, Warscheid B. Maintaining proteostasis under mechanical stress. EMBO Rep 2021; 22:e52507. [PMID: 34309183 PMCID: PMC8339670 DOI: 10.15252/embr.202152507] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
Cell survival, tissue integrity and organismal health depend on the ability to maintain functional protein networks even under conditions that threaten protein integrity. Protection against such stress conditions involves the adaptation of folding and degradation machineries, which help to preserve the protein network by facilitating the refolding or disposal of damaged proteins. In multicellular organisms, cells are permanently exposed to stress resulting from mechanical forces. Yet, for long time mechanical stress was not recognized as a primary stressor that perturbs protein structure and threatens proteome integrity. The identification and characterization of protein folding and degradation systems, which handle force-unfolded proteins, marks a turning point in this regard. It has become apparent that mechanical stress protection operates during cell differentiation, adhesion and migration and is essential for maintaining tissues such as skeletal muscle, heart and kidney as well as the immune system. Here, we provide an overview of recent advances in our understanding of mechanical stress protection.
Collapse
Affiliation(s)
- Jörg Höhfeld
- Institute for Cell BiologyRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sports MedicineGerman Sport UniversityCologneGermany
| | - Dieter O Fürst
- Institute for Cell BiologyRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Sebastian Gehlert
- Institute of Cardiovascular Research and Sports MedicineGerman Sport UniversityCologneGermany
- Department for the Biosciences of SportsInstitute of Sports ScienceUniversity of HildesheimHildesheimGermany
| | - Michael Hesse
- Institute of Physiology I, Life & Brain CenterMedical FacultyRheinische Friedrich‐Wilhelms UniversityBonnGermany
| | - Bernd Hoffmann
- Institute of Biological Information Processing, IBI‐2: MechanobiologyForschungszentrum JülichJülichGermany
| | - Thorsten Hoppe
- Institute for GeneticsCologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD) and CMMCUniversity of CologneCologneGermany
| | - Pitter F Huesgen
- Central Institute for Engineering, Electronics and Analytics, ZEA3Forschungszentrum JülichJülichGermany
- CECADUniversity of CologneCologneGermany
| | - Maja Köhn
- Institute of Biology IIIFaculty of Biology, and Signalling Research Centres BIOSS and CIBSSAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Waldemar Kolanus
- LIMES‐InstituteRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Rudolf Merkel
- Institute of Biological Information Processing, IBI‐2: MechanobiologyForschungszentrum JülichJülichGermany
| | - Carien M Niessen
- Department of Dermatology and CECADUniversity of CologneCologneGermany
| | | | - Markus M Rinschen
- Department of Biomedicine and Aarhus Institute of Advanced StudiesAarhus UniversityAarhusDenmark
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Dagmar Wachten
- Institute of Innate ImmunityUniversity Hospital BonnBonnGermany
| | - Bettina Warscheid
- Institute of Biology IIFaculty of Biology, and Signalling Research Centres BIOSS and CIBSSAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| |
Collapse
|
18
|
Chen X, Yin J, Cao D, Xiao D, Zhou Z, Liu Y, Shou W. The Emerging Roles of the RNA Binding Protein QKI in Cardiovascular Development and Function. Front Cell Dev Biol 2021; 9:668659. [PMID: 34222237 PMCID: PMC8242579 DOI: 10.3389/fcell.2021.668659] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/10/2021] [Indexed: 12/30/2022] Open
Abstract
RNA binding proteins (RBPs) have a broad biological and physiological function and are critical in regulating pre-mRNA posttranscriptional processing, intracellular migration, and mRNA stability. QKI, also known as Quaking, is a member of the signal transduction and activation of RNA (STAR) family, which also belongs to the heterogeneous nuclear ribonucleoprotein K- (hnRNP K-) homology domain protein family. There are three major alternatively spliced isoforms, QKI-5, QKI-6, and QKI-7, differing in carboxy-terminal domains. They share a common RNA binding property, but each isoform can regulate pre-mRNA splicing, transportation or stability differently in a unique cell type-specific manner. Previously, QKI has been known for its important role in contributing to neurological disorders. A series of recent work has further demonstrated that QKI has important roles in much broader biological systems, such as cardiovascular development, monocyte to macrophage differentiation, bone metabolism, and cancer progression. In this mini-review, we will focus on discussing the emerging roles of QKI in regulating cardiac and vascular development and function and its potential link to cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Xinyun Chen
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen, China
| | - Jianwen Yin
- Department of Foot, Ankle and Hand Surgery, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Dayan Cao
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Deyong Xiao
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Zhongjun Zhou
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Hong Kong
| | - Ying Liu
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Weinian Shou
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
19
|
Müller D, Klamt T, Gentemann L, Heisterkamp A, Kalies SMK. Evaluation of laser induced sarcomere micro-damage: Role of damage extent and location in cardiomyocytes. PLoS One 2021; 16:e0252346. [PMID: 34086732 PMCID: PMC8177425 DOI: 10.1371/journal.pone.0252346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/12/2021] [Indexed: 11/18/2022] Open
Abstract
Whereas it is evident that a well aligned and regular sarcomeric structure in cardiomyocytes is vital for heart function, considerably less is known about the contribution of individual elements to the mechanics of the entire cell. For instance, it is unclear whether altered Z-disc elements are the reason or the outcome of related cardiomyopathies. Therefore, it is crucial to gain more insight into this cellular organization. This study utilizes femtosecond laser-based nanosurgery to better understand sarcomeres and their repair upon damage. We investigated the influence of the extent and the location of the Z-disc damage. A single, three, five or ten Z-disc ablations were performed in neonatal rat cardiomyocytes. We employed image-based analysis using a self-written software together with different already published algorithms. We observed that cardiomyocyte survival associated with the damage extent, but not with the cell area or the total number of Z-discs per cell. The cell survival is independent of the damage position and can be compensated. However, the sarcomere alignment/orientation is changing over time after ablation. The contraction time is also independent of the extent of damage for the tested parameters. Additionally, we observed shortening rates between 6–7% of the initial sarcomere length in laser treated cardiomyocytes. This rate is an important indicator for force generation in myocytes. In conclusion, femtosecond laser-based nanosurgery together with image-based sarcomere tracking is a powerful tool to better understand the Z-disc complex and its force propagation function and role in cellular mechanisms.
Collapse
Affiliation(s)
- Dominik Müller
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
- * E-mail:
| | - Thorben Klamt
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Lara Gentemann
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Stefan Michael Klaus Kalies
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| |
Collapse
|
20
|
Sponga A, Arolas JL, Schwarz TC, Jeffries CM, Rodriguez Chamorro A, Kostan J, Ghisleni A, Drepper F, Polyansky A, De Almeida Ribeiro E, Pedron M, Zawadzka-Kazimierczuk A, Mlynek G, Peterbauer T, Doto P, Schreiner C, Hollerl E, Mateos B, Geist L, Faulkner G, Kozminski W, Svergun DI, Warscheid B, Zagrovic B, Gautel M, Konrat R, Djinović-Carugo K. Order from disorder in the sarcomere: FATZ forms a fuzzy but tight complex and phase-separated condensates with α-actinin. SCIENCE ADVANCES 2021; 7:eabg7653. [PMID: 34049882 PMCID: PMC8163081 DOI: 10.1126/sciadv.abg7653] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/13/2021] [Indexed: 05/03/2023]
Abstract
In sarcomeres, α-actinin cross-links actin filaments and anchors them to the Z-disk. FATZ (filamin-, α-actinin-, and telethonin-binding protein of the Z-disk) proteins interact with α-actinin and other core Z-disk proteins, contributing to myofibril assembly and maintenance. Here, we report the first structure and its cellular validation of α-actinin-2 in complex with a Z-disk partner, FATZ-1, which is best described as a conformational ensemble. We show that FATZ-1 forms a tight fuzzy complex with α-actinin-2 and propose an interaction mechanism via main molecular recognition elements and secondary binding sites. The obtained integrative model reveals a polar architecture of the complex which, in combination with FATZ-1 multivalent scaffold function, might organize interaction partners and stabilize α-actinin-2 preferential orientation in Z-disk. Last, we uncover FATZ-1 ability to phase-separate and form biomolecular condensates with α-actinin-2, raising the question whether FATZ proteins can create an interaction hub for Z-disk proteins through membraneless compartmentalization during myofibrillogenesis.
Collapse
Affiliation(s)
- Antonio Sponga
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Joan L Arolas
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Thomas C Schwarz
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Cy M Jeffries
- European Molecular Biology Laboratory (EMBL), Hamburg Unit, Hamburg, Germany
| | - Ariadna Rodriguez Chamorro
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Julius Kostan
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Andrea Ghisleni
- King's College London BHF Centre for Research Excellence, Randall Centre for Cell and Molecular Biophysics, London SE1 1UL, UK
| | - Friedel Drepper
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Anton Polyansky
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
- National Research University Higher School of Economics, Moscow 101000, Russia
| | - Euripedes De Almeida Ribeiro
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Miriam Pedron
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Anna Zawadzka-Kazimierczuk
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, Zwirki i Wigury 101, 02-089 Warsaw, Poland
| | - Georg Mlynek
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Thomas Peterbauer
- Department of Biochemistry and Cell Biology, Max Perutz Labs, University of Vienna, Dr. BohrGasse 9, A-1030 Vienna, Austria
| | - Pierantonio Doto
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Claudia Schreiner
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Eneda Hollerl
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Borja Mateos
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Leonhard Geist
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | | | - Wiktor Kozminski
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, Zwirki i Wigury 101, 02-089 Warsaw, Poland
| | - Dmitri I Svergun
- King's College London BHF Centre for Research Excellence, Randall Centre for Cell and Molecular Biophysics, London SE1 1UL, UK
| | - Bettina Warscheid
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Bojan Zagrovic
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Mathias Gautel
- King's College London BHF Centre for Research Excellence, Randall Centre for Cell and Molecular Biophysics, London SE1 1UL, UK
| | - Robert Konrat
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Kristina Djinović-Carugo
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria.
- Department of Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
21
|
Riechert E, Kmietczyk V, Stein F, Schwarzl T, Sekaran T, Jürgensen L, Kamuf-Schenk V, Varma E, Hofmann C, Rettel M, Gür K, Ölschläger J, Kühl F, Martin J, Ramirez-Pedraza M, Fernandez M, Doroudgar S, Méndez R, Katus HA, Hentze MW, Völkers M. Identification of dynamic RNA-binding proteins uncovers a Cpeb4-controlled regulatory cascade during pathological cell growth of cardiomyocytes. Cell Rep 2021; 35:109100. [PMID: 33979607 DOI: 10.1016/j.celrep.2021.109100] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 02/03/2021] [Accepted: 04/16/2021] [Indexed: 11/18/2022] Open
Abstract
RNA-binding proteins (RBPs) control critical aspects of cardiomyocyte function, but the repertoire of active RBPs in cardiomyocytes during the growth response is largely unknown. We define RBPs in healthy and diseased cardiomyocytes at a system-wide level by RNA interactome capture. This identifies 67 cardiomyocyte-specific RBPs, including several contractile proteins. Furthermore, we identify the cytoplasmic polyadenylation element-binding protein 4 (Cpeb4) as a dynamic RBP, regulating cardiac growth both in vitro and in vivo. We identify mRNAs bound to and regulated by Cpeb4 in cardiomyocytes. Cpeb4 regulates cardiac remodeling by differential expression of transcription factors. Among Cpeb4 target mRNAs, two zinc finger transcription factors (Zeb1 and Zbtb20) are discovered. We show that Cpeb4 regulates the expression of these mRNAs and that Cpeb4 depletion increases their expression. Thus, Cpeb4 emerges as a critical regulator of cardiomyocyte function by differential binding to specific mRNAs in response to pathological growth stimulation.
Collapse
Affiliation(s)
- Eva Riechert
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Vivien Kmietczyk
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Thomas Schwarzl
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Thileepan Sekaran
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Lonny Jürgensen
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Verena Kamuf-Schenk
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Eshita Varma
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Christoph Hofmann
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Mandy Rettel
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Kira Gür
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Julie Ölschläger
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Friederike Kühl
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Judit Martin
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | | | | | - Shirin Doroudgar
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Raúl Méndez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; Institució Catalana de Recerca I Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Hugo A Katus
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Matthias W Hentze
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Mirko Völkers
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany.
| |
Collapse
|
22
|
Maintenance of type 2 glycolytic myofibers with age by Mib1-Actn3 axis. Nat Commun 2021; 12:1294. [PMID: 33637766 PMCID: PMC7910585 DOI: 10.1038/s41467-021-21621-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023] Open
Abstract
Age-associated muscle atrophy is a debilitating condition associated with loss of muscle mass and function with age that contributes to limitation of mobility and locomotion. However, the underlying mechanisms of how intrinsic muscle changes with age are largely unknown. Here we report that, with age, Mind bomb-1 (Mib1) plays important role in skeletal muscle maintenance via proteasomal degradation-dependent regulation of α-actinin 3 (Actn3). The disruption of Mib1 in myofibers (Mib1ΔMF) results in alteration of type 2 glycolytic myofibers, muscle atrophy, impaired muscle function, and Actn3 accumulation. After chronic exercise, Mib1ΔMF mice show muscle atrophy even at young age. However, when Actn3 level is downregulated, chronic exercise-induced muscle atrophy is ameliorated. Importantly, the Mib1 and Actn3 levels show clinical relevance in human skeletal muscles accompanied by decrease in skeletal muscle function with age. Together, these findings reveal the significance of the Mib1-Actn3 axis in skeletal muscle maintenance with age and suggest the therapeutic potential for the treatment or amelioration of age-related muscle atrophy.
Collapse
|
23
|
Amrute‐Nayak M, Pegoli G, Holler T, Lopez‐Davila AJ, Lanzuolo C, Nayak A. Chemotherapy triggers cachexia by deregulating synergetic function of histone-modifying enzymes. J Cachexia Sarcopenia Muscle 2021; 12:159-176. [PMID: 33305533 PMCID: PMC7890149 DOI: 10.1002/jcsm.12645] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 08/10/2020] [Accepted: 10/12/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Chemotherapy is the first line of treatment for cancer patients. However, the side effects cause severe muscle atrophy or chemotherapy-induced cachexia. Previously, the NF-κB/MuRF1-dependent pathway was shown to induce chemotherapy-induced cachexia. We hypothesized that acute collateral toxic effects of chemotherapy on muscles might involve other unknown pathways promoting chemotherapy-induced muscle atrophy. In this study, we investigated differential effects of chemotherapeutic drugs and probed whether alternative molecular mechanisms lead to cachexia. METHODS We employed mouse satellite stem cell-derived primary muscle cells and mouse C2C12 progenitor cell-derived differentiated myotubes as model systems to test the effect of drugs. The widely used chemotherapeutic drugs, such as daunorubicin (Daun), etoposide (Etop), and cytarabine (Ara-C), were tested. Molecular mechanisms by which drug affects the muscle cell organization at epigenetic, transcriptional, and protein levels were measured by employing chromatin immunoprecipitations, endogenous gene expression profiling, co-immunoprecipitation, complementation assays, and confocal microscopy. Myotube function was examined using the electrical stimulation of myotubes to monitor contractile ability (excitation-contraction coupling) post drug treatment. RESULTS Here, we demonstrate that chemotherapeutic drugs disrupt sarcomere organization and thereby the contractile ability of skeletal muscle cells. The sarcomere disorganization results from severe loss of molecular motor protein MyHC-II upon drug treatment. We identified that drugs impede chromatin targeting of SETD7 histone methyltransferase and disrupt association and synergetic function of SETD7 with p300 histone acetyltransferase. The compromised transcriptional activity of histone methyltransferase and acetyltransferase causes reduced histone acetylation and low occupancy of active RNA polymerase II on MyHC-II, promoting drastic down-regulation of MyHC-II expression (~3.6-fold and ~4.5-fold reduction of MyHC-IId mRNA levels in Daun and Etop treatment, respectively. P < 0.0001). For MyHC-IIa, gene expression was down-regulated by ~2.6-fold and ~4.5-fold in Daun and Etop treatment, respectively (P < 0.0001). Very interestingly, the drugs destabilize SUMO deconjugase SENP3. Reduction in SENP3 protein level leads to deregulation of SETD7-p300 function. Importantly, we identified that SUMO deconjugation independent role of SENP3 regulates SETD7-p300 functional axis. CONCLUSIONS The results show that the drugs critically alter SENP3-dependent synergistic action of histone-modifying enzymes in muscle cells. Collectively, we defined a unique epigenetic mechanism targeted by distinct chemotherapeutic drugs, triggering chemotherapy-induced cachexia.
Collapse
Affiliation(s)
- Mamta Amrute‐Nayak
- Institute of Molecular and Cell PhysiologyHannover Medical SchoolHannoverGermany
| | - Gloria Pegoli
- Institute of Biomedical TechnologiesNational Research CouncilMilanItaly
- Istituto Nazionale Genetica Molecolare ‘Romeo ed Enrica Invernizzi’MilanItaly
| | - Tim Holler
- Institute of Molecular and Cell PhysiologyHannover Medical SchoolHannoverGermany
| | | | - Chiara Lanzuolo
- Institute of Biomedical TechnologiesNational Research CouncilMilanItaly
- Istituto Nazionale Genetica Molecolare ‘Romeo ed Enrica Invernizzi’MilanItaly
| | - Arnab Nayak
- Institute of Molecular and Cell PhysiologyHannover Medical SchoolHannoverGermany
| |
Collapse
|
24
|
Ramaccini D, Montoya-Uribe V, Aan FJ, Modesti L, Potes Y, Wieckowski MR, Krga I, Glibetić M, Pinton P, Giorgi C, Matter ML. Mitochondrial Function and Dysfunction in Dilated Cardiomyopathy. Front Cell Dev Biol 2021; 8:624216. [PMID: 33511136 PMCID: PMC7835522 DOI: 10.3389/fcell.2020.624216] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiac tissue requires a persistent production of energy in order to exert its pumping function. Therefore, the maintenance of this function relies on mitochondria that represent the “powerhouse” of all cardiac activities. Mitochondria being one of the key players for the proper functioning of the mammalian heart suggests continual regulation and organization. Mitochondria adapt to cellular energy demands via fusion-fission events and, as a proof-reading ability, undergo mitophagy in cases of abnormalities. Ca2+ fluxes play a pivotal role in regulating all mitochondrial functions, including ATP production, metabolism, oxidative stress balance and apoptosis. Communication between mitochondria and others organelles, especially the sarcoplasmic reticulum is required for optimal function. Consequently, abnormal mitochondrial activity results in decreased energy production leading to pathological conditions. In this review, we will describe how mitochondrial function or dysfunction impacts cardiac activities and the development of dilated cardiomyopathy.
Collapse
Affiliation(s)
- Daniela Ramaccini
- University of Hawaii Cancer Center, Honolulu, HI, United States.,Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy
| | | | - Femke J Aan
- University of Hawaii Cancer Center, Honolulu, HI, United States
| | - Lorenzo Modesti
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy
| | - Yaiza Potes
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Irena Krga
- Center of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Marija Glibetić
- Center of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy.,Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy
| | | |
Collapse
|
25
|
Czajkowski ER, Cisneros M, Garcia BS, Shen J, Cripps RM. The Drosophila CG1674 gene encodes a synaptopodin 2-like related protein that localizes to the Z-disc and is required for normal flight muscle development and function. Dev Dyn 2021; 250:99-110. [PMID: 32893414 PMCID: PMC7902442 DOI: 10.1002/dvdy.250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/12/2020] [Accepted: 09/01/2020] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND To identify novel myofibrillar components of the Drosophila flight muscles, we carried out a proteomic analysis of chemically demembranated flight muscle myofibrils, and characterized the knockdown phenotype of a novel gene identified in the screen, CG1674. RESULTS The CG1674 protein has some similarity to vertebrate synaptopodin 2-like, and when expressed as a FLAG-tagged fusion protein, it was localized during development to the Z-disc and cytoplasm. Knockdown of CG1674 expression affected the function of multiple muscle types, and defective flight in adults was accompanied by large actin-rich structures in the flight muscles that resembled overgrown Z-discs. Localization of CG1674 to the Z-disc depended predominantly upon presence of the Z-disc component alpha-actinin, but also depended upon other Z-disc components, including Mask, Zasp52, and Sals. We also observed re-localization of FLAG-CG1674 to the nucleus in Alpha-actinin and sals knockdown animals. CONCLUSIONS These studies identify and characterize a previously unreported myofibrillar component of Drosophila muscle that is necessary for proper myofibril assembly during development.
Collapse
Affiliation(s)
| | - Marilyn Cisneros
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Bianca S. Garcia
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Jim Shen
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Richard M. Cripps
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| |
Collapse
|
26
|
Vikhorev PG, Vikhoreva NN, Yeung W, Li A, Lal S, dos Remedios CG, Blair CA, Guglin M, Campbell KS, Yacoub MH, de Tombe P, Marston SB. Titin-truncating mutations associated with dilated cardiomyopathy alter length-dependent activation and its modulation via phosphorylation. Cardiovasc Res 2020; 118:241-253. [PMID: 33135063 PMCID: PMC8752363 DOI: 10.1093/cvr/cvaa316] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 10/20/2020] [Indexed: 12/18/2022] Open
Abstract
Abstract
Aims
Dilated cardiomyopathy (DCM) is associated with mutations in many genes encoding sarcomere proteins. Truncating mutations in the titin gene TTN are the most frequent. Proteomic and functional characterizations are required to elucidate the origin of the disease and the pathogenic mechanisms of TTN-truncating variants.
Methods and results
We isolated myofibrils from DCM hearts carrying truncating TTN mutations and measured the Ca2+ sensitivity of force and its length dependence. Simultaneous measurement of force and adenosine triphosphate (ATP) consumption in skinned cardiomyocytes was also performed. Phosphorylation levels of troponin I (TnI) and myosin binding protein-C (MyBP-C) were manipulated using protein kinase A and λ phosphatase. mRNA sequencing was employed to overview gene expression profiles. We found that Ca2+ sensitivity of myofibrils carrying TTN mutations was significantly higher than in myofibrils from donor hearts. The length dependence of the Ca2+ sensitivity was absent in DCM myofibrils with TTN-truncating variants. No significant difference was found in the expression level of TTN mRNA between the DCM and donor groups. TTN exon usage and splicing were also similar. However, we identified down-regulation of genes encoding Z-disk proteins, while the atrial-specific regulatory myosin light chain gene, MYL7, was up-regulated in DCM patients with TTN-truncating variants.
Conclusion
Titin-truncating mutations lead to decreased length-dependent activation and increased elasticity of myofibrils. Phosphorylation levels of TnI and MyBP-C seen in the left ventricles are essential for the length-dependent changes in Ca2+ sensitivity in healthy donors, but they are reduced in DCM patients with TTN-truncating variants. A decrease in expression of Z-disk proteins may explain the observed decrease in myofibril passive stiffness and length-dependent activation.
Collapse
Affiliation(s)
- Petr G Vikhorev
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Natalia N Vikhoreva
- Heart Science Centre, Magdi Yacoub Institute, Harefield Hospital, London UB9 6JH, UK
| | - WaiChun Yeung
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Amy Li
- Department of Pharmacy and Biomedical Sciences, La Trobe University, Bendigo, VIC 3550, Australia
| | - Sean Lal
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Cristobal G dos Remedios
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Cheavar A Blair
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Maya Guglin
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Kenneth S Campbell
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Magdi H Yacoub
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Pieter de Tombe
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
- Heart Science Centre, Magdi Yacoub Institute, Harefield Hospital, London UB9 6JH, UK
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Steven B Marston
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
27
|
Distinct hypertrophic cardiomyopathy genotypes result in convergent sarcomeric proteoform profiles revealed by top-down proteomics. Proc Natl Acad Sci U S A 2020; 117:24691-24700. [PMID: 32968017 PMCID: PMC7547245 DOI: 10.1073/pnas.2006764117] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common heritable heart disease. Although the genetic cause of HCM has been linked to mutations in genes encoding sarcomeric proteins, the ability to predict clinical outcomes based on specific mutations in HCM patients is limited. Moreover, how mutations in different sarcomeric proteins can result in highly similar clinical phenotypes remains unknown. Posttranslational modifications (PTMs) and alternative splicing regulate the function of sarcomeric proteins; hence, it is critical to study HCM at the level of proteoforms to gain insights into the mechanisms underlying HCM. Herein, we employed high-resolution mass spectrometry-based top-down proteomics to comprehensively characterize sarcomeric proteoforms in septal myectomy tissues from HCM patients exhibiting severe outflow track obstruction (n = 16) compared to nonfailing donor hearts (n = 16). We observed a complex landscape of sarcomeric proteoforms arising from combinatorial PTMs, alternative splicing, and genetic variation in HCM. A coordinated decrease of phosphorylation in important myofilament and Z-disk proteins with a linear correlation suggests PTM cross-talk in the sarcomere and dysregulation of protein kinase A pathways in HCM. Strikingly, we discovered that the sarcomeric proteoform alterations in the myocardium of HCM patients undergoing septal myectomy were remarkably consistent, regardless of the underlying HCM-causing mutations. This study suggests that the manifestation of severe HCM coalesces at the proteoform level despite distinct genotype, which underscores the importance of molecular characterization of HCM phenotype and presents an opportunity to identify broad-spectrum treatments to mitigate the most severe manifestations of this genetically heterogenous disease.
Collapse
|
28
|
Toste A, Perrot A, Özcelik C, Cardim N. Identification of a novel titin-cap/telethonin mutation in a Portuguese family with hypertrophic cardiomyopathy. Rev Port Cardiol 2020; 39:317-327. [PMID: 32565061 DOI: 10.1016/j.repc.2019.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/04/2019] [Accepted: 12/19/2019] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION AND OBJECTIVES Hypertrophic cardiomyopathy (HCM) is a genetically and phenotypically heterogeneous disease; there is still a large proportion of patients with no identified disease-causing mutation. Although the majority of mutations are found in the MYH7 and MYBPC3 genes, mutations in Z-disk-associated proteins have also been linked to HCM. METHODS We assessed a small family with HCM based on family history, physical examination, 12-lead ECG, echocardiogram and magnetic resonance imaging. After exclusion of mutations in eleven HCM disease genes, we performed direct sequencing of the TCAP gene encoding the Z-disk protein titin-cap (also known as telethonin). RESULTS We present a novel TCAP mutation in a small family affected by HCM. The identified p.C57W mutation showed a very low population frequency, as well as high conservation across species. All of the bioinformatic prediction tools used considered this mutation to be damaging/deleterious. Family members were screened for this new mutation and a co-segregation pattern was detected. Both affected members of this family presented with late-onset HCM, moderate asymmetric left ventricular hypertrophy, atrial fibrillation and heart failure with preserved ejection fraction and low risk of sudden cardiac death. CONCLUSIONS We present evidence supporting the classification of the TCAP p.C57W mutation, encoding the Z-disk protein titin-cap/telethonin as a new likely pathogenic variant of hypertrophic cardiomyopathy, with a specific phenotype in the family under analysis.
Collapse
Affiliation(s)
- Alexandra Toste
- Hospital da Luz - Inherited Cardiovascular Diseases & Hypertrophic Cardiomyopathy Center, Nova Medical School, Lisbon, Portugal.
| | - Andreas Perrot
- Charité-Universitätsmedizin Berlin, Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Cemil Özcelik
- Helios Klinikum Emil von Behring GmbH, Department of Internal Medicine - Cardiology, Berlin, Germany
| | - Nuno Cardim
- Hospital da Luz - Inherited Cardiovascular Diseases & Hypertrophic Cardiomyopathy Center, Nova Medical School, Lisbon, Portugal
| |
Collapse
|
29
|
Toste A, Perrot A, Özcelik C, Cardim N. Identification of a novel titin-cap/telethonin mutation in a Portuguese family with hypertrophic cardiomyopathy. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2020. [DOI: 10.1016/j.repce.2019.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
30
|
Role of HDACs in cardiac electropathology: Therapeutic implications for atrial fibrillation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118459. [DOI: 10.1016/j.bbamcr.2019.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 12/21/2022]
|
31
|
Lambert M, Claeyssen C, Bastide B, Cieniewski‐Bernard C. O-GlcNAcylation as a regulator of the functional and structural properties of the sarcomere in skeletal muscle: An update review. Acta Physiol (Oxf) 2020; 228:e13301. [PMID: 31108020 DOI: 10.1111/apha.13301] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/03/2019] [Accepted: 05/10/2019] [Indexed: 12/15/2022]
Abstract
Although the O-GlcNAcylation process was discovered in 1984, its potential role in the physiology and physiopathology of skeletal muscle only emerged 20 years later. An increasing number of publications strongly support a key role of O-GlcNAcylation in the modulation of important cellular processes which are essential for skeletal muscle functions. Indeed, over a thousand of O-GlcNAcylated proteins have been identified within skeletal muscle since 2004, which belong to various classes of proteins, including sarcomeric proteins. In this review, we focused on these myofibrillar proteins, including contractile and structural proteins. Because of the modification of motor and regulatory proteins, the regulatory myosin light chain (MLC2) is related to several reports that support a key role of O-GlcNAcylation in the fine modulation of calcium activation parameters of skeletal muscle fibres, depending on muscle phenotype and muscle work. In addition, another key function of O-GlcNAcylation has recently emerged in the regulation of organization and reorganization of the sarcomere. Altogether, this data support a key role of O-GlcNAcylation in the homeostasis of sarcomeric cytoskeleton, known to be disturbed in many related muscle disorders.
Collapse
Affiliation(s)
- Matthias Lambert
- Univ. Lille, EA 7369 ‐ URePSSS ‐ Unité de Recherche Pluridisciplinaire Sport Santé Société Lille France
| | - Charlotte Claeyssen
- Univ. Lille, EA 7369 ‐ URePSSS ‐ Unité de Recherche Pluridisciplinaire Sport Santé Société Lille France
| | - Bruno Bastide
- Univ. Lille, EA 7369 ‐ URePSSS ‐ Unité de Recherche Pluridisciplinaire Sport Santé Société Lille France
| | | |
Collapse
|
32
|
Deficiency of nuclear receptor interaction protein leads to cardiomyopathy by disrupting sarcomere structure and mitochondrial respiration. J Mol Cell Cardiol 2019; 137:9-24. [PMID: 31629737 DOI: 10.1016/j.yjmcc.2019.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/15/2019] [Accepted: 09/17/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Cardiomyopathy is a common and lethal complication in patients with limb-girdle muscular dystrophy (LGMD), one of the most prevalent forms of muscular dystrophy. The pathogenesis underlying LGMD-related cardiomyopathy remains unclear. NRIP (gene name DCAF6), a Ca2+-dependent calmodulin binding protein, was reduced in dystrophic muscles from LGMD patients. Mice lacking NRIP exhibit a myopathic phenotype resembling that in LGMD patients, making NRIP deficiency a potential culprit leading to cardiomyopathy. This study aimed to determine if NRIP deficiency leads to cardiomyopathy and to explore the underlying molecular mechanisms. METHODS AND RESULTS NRIP expression was reduced in both human and mouse failing hearts. Muscle-specific NRIP knockout (MCK-Cre::Dcaf6flox/flox) mouse heart and isolated cardiomyocytes exhibited markedly reduced contractility. Transmission electron microscopy revealed abnormal sarcomere structures and mitochondrial morphology in MCK-Cre::Dcaf6flox/flox hearts. Protein co-immunoprecipitation and confocal imaging revealed that NRIP interacts with α-actinin 2 (ACTN2) at the Z-disc. We found that NRIP facilitated ACTN2-mediated F-actin bundling, and that NRIP deficiency resulted in reduced binding between Z-disc proteins ACTN2 and Cap-Z. In addition, NRIP-deficiency led to increased mitochondrial ROS and impaired mitochondrial respiration/ATP production owing to elevated cellular NADH/NAD+ ratios. Treatment with mitochondria-directed antioxidant mitoTEMPO or NAD+ precursor nicotinic acid restored mitochondrial function and cardiac contractility in MCK-Cre::Dcaf6flox/flox mice. CONCLUSIONS NRIP is essential to maintain sarcomere structure and mitochondrial/contractile function in cardiomyocytes. Our results revealed a novel role for NRIP deficiency in the pathogenesis of LGMD and heart failure. Targeting NRIP, therefore, could be a powerful new approach to treat myocardial dysfunction in LGMD and heart failure patients.
Collapse
|
33
|
Good JM, Fellmann F, Bhuiyan ZA, Rotman S, Pruvot E, Schläpfer J. ACTN2 variant associated with a cardiac phenotype suggestive of left-dominant arrhythmogenic cardiomyopathy. HeartRhythm Case Rep 2019; 6:15-19. [PMID: 31956495 PMCID: PMC6962717 DOI: 10.1016/j.hrcr.2019.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Jean-Marc Good
- Department of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Florence Fellmann
- Department of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Zahurul A Bhuiyan
- Department of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Samuel Rotman
- Service of Clinical Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Etienne Pruvot
- Department of Cardiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jürg Schläpfer
- Department of Cardiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
34
|
Burgoyne T, Heumann JM, Morris EP, Knupp C, Liu J, Reedy MK, Taylor KA, Wang K, Luther PK. Three-dimensional structure of the basketweave Z-band in midshipman fish sonic muscle. Proc Natl Acad Sci U S A 2019; 116:15534-15539. [PMID: 31320587 PMCID: PMC6681754 DOI: 10.1073/pnas.1902235116] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Striated muscle enables movement in all animals by the contraction of myriads of sarcomeres joined end to end by the Z-bands. The contraction is due to tension generated in each sarcomere between overlapping arrays of actin and myosin filaments. At the Z-band, actin filaments from adjoining sarcomeres overlap and are cross-linked in a regular pattern mainly by the protein α-actinin. The Z-band is dynamic, reflected by the 2 regular patterns seen in transverse section electron micrographs; the so-called small-square and basketweave forms. Although these forms are attributed, respectively, to relaxed and actively contracting muscles, the basketweave form occurs in certain relaxed muscles as in the muscle studied here. We used electron tomography and subtomogram averaging to derive the 3D structure of the Z-band in the swimbladder sonic muscle of type I male plainfin midshipman fish (Porichthys notatus), into which we docked the crystallographic structures of actin and α-actinin. The α-actinin links run diagonally between connected pairs of antiparallel actin filaments and are oriented at an angle of about 25° away from the actin filament axes. The slightly curved and flattened structure of the α-actinin rod has a distinct fit into the map. The Z-band model provides a detailed understanding of the role of α-actinin in transmitting tension between actin filaments in adjoining sarcomeres.
Collapse
Affiliation(s)
- Thomas Burgoyne
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, SW7 2AZ London, United Kingdom
| | - John M Heumann
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309-0347
| | - Edward P Morris
- Division of Structural Biology, Institute of Cancer Research, SW7 3RP London, United Kingdom
| | - Carlo Knupp
- School of Optometry and Vision Sciences, Cardiff University, CF10 3AT Cardiff, United Kingdom
| | - Jun Liu
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306-4380
| | - Michael K Reedy
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710
| | - Kenneth A Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306-4380
| | - Kuan Wang
- Laboratory of Muscle Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892
- College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Pradeep K Luther
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, SW7 2AZ London, United Kingdom;
| |
Collapse
|
35
|
Towbin JA, McKenna WJ, Abrams DJ, Ackerman MJ, Calkins H, Darrieux FCC, Daubert JP, de Chillou C, DePasquale EC, Desai MY, Estes NAM, Hua W, Indik JH, Ingles J, James CA, John RM, Judge DP, Keegan R, Krahn AD, Link MS, Marcus FI, McLeod CJ, Mestroni L, Priori SG, Saffitz JE, Sanatani S, Shimizu W, van Tintelen JP, Wilde AAM, Zareba W. 2019 HRS expert consensus statement on evaluation, risk stratification, and management of arrhythmogenic cardiomyopathy. Heart Rhythm 2019; 16:e301-e372. [PMID: 31078652 DOI: 10.1016/j.hrthm.2019.05.007] [Citation(s) in RCA: 454] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Indexed: 02/08/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an arrhythmogenic disorder of the myocardium not secondary to ischemic, hypertensive, or valvular heart disease. ACM incorporates a broad spectrum of genetic, systemic, infectious, and inflammatory disorders. This designation includes, but is not limited to, arrhythmogenic right/left ventricular cardiomyopathy, cardiac amyloidosis, sarcoidosis, Chagas disease, and left ventricular noncompaction. The ACM phenotype overlaps with other cardiomyopathies, particularly dilated cardiomyopathy with arrhythmia presentation that may be associated with ventricular dilatation and/or impaired systolic function. This expert consensus statement provides the clinician with guidance on evaluation and management of ACM and includes clinically relevant information on genetics and disease mechanisms. PICO questions were utilized to evaluate contemporary evidence and provide clinical guidance related to exercise in arrhythmogenic right ventricular cardiomyopathy. Recommendations were developed and approved by an expert writing group, after a systematic literature search with evidence tables, and discussion of their own clinical experience, to present the current knowledge in the field. Each recommendation is presented using the Class of Recommendation and Level of Evidence system formulated by the American College of Cardiology and the American Heart Association and is accompanied by references and explanatory text to provide essential context. The ongoing recognition of the genetic basis of ACM provides the opportunity to examine the diverse triggers and potential common pathway for the development of disease and arrhythmia.
Collapse
Affiliation(s)
- Jeffrey A Towbin
- Le Bonheur Children's Hospital, Memphis, Tennessee; University of Tennessee Health Science Center, Memphis, Tennessee
| | - William J McKenna
- University College London, Institute of Cardiovascular Science, London, United Kingdom
| | | | | | | | | | | | | | | | | | - N A Mark Estes
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Wei Hua
- Fu Wai Hospital, Beijing, China
| | - Julia H Indik
- University of Arizona, Sarver Heart Center, Tucson, Arizona
| | - Jodie Ingles
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, Australia
| | | | - Roy M John
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Daniel P Judge
- Medical University of South Carolina, Charleston, South Carolina
| | - Roberto Keegan
- Hospital Privado Del Sur, Buenos Aires, Argentina; Hospital Español, Bahia Blanca, Argentina
| | | | - Mark S Link
- UT Southwestern Medical Center, Dallas, Texas
| | - Frank I Marcus
- University of Arizona, Sarver Heart Center, Tucson, Arizona
| | | | - Luisa Mestroni
- University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Silvia G Priori
- University of Pavia, Pavia, Italy; European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-Heart); ICS Maugeri, IRCCS, Pavia, Italy
| | | | | | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | - J Peter van Tintelen
- University of Amsterdam, Academic Medical Center, Amsterdam, the Netherlands; Utrecht University Medical Center Utrecht, University of Utrecht, Department of Genetics, Utrecht, the Netherlands
| | - Arthur A M Wilde
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-Heart); University of Amsterdam, Academic Medical Center, Amsterdam, the Netherlands; Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | | |
Collapse
|
36
|
Li Y, Merkel CD, Zeng X, Heier JA, Cantrell PS, Sun M, Stolz DB, Watkins SC, Yates NA, Kwiatkowski AV. The N-cadherin interactome in primary cardiomyocytes as defined using quantitative proximity proteomics. J Cell Sci 2019; 132:jcs.221606. [PMID: 30630894 PMCID: PMC6382013 DOI: 10.1242/jcs.221606] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 12/24/2018] [Indexed: 12/11/2022] Open
Abstract
The junctional complexes that couple cardiomyocytes must transmit the mechanical forces of contraction while maintaining adhesive homeostasis. The adherens junction (AJ) connects the actomyosin networks of neighboring cardiomyocytes and is required for proper heart function. Yet little is known about the molecular composition of the cardiomyocyte AJ or how it is organized to function under mechanical load. Here, we define the architecture, dynamics and proteome of the cardiomyocyte AJ. Mouse neonatal cardiomyocytes assemble stable AJs along intercellular contacts with organizational and structural hallmarks similar to mature contacts. We combine quantitative mass spectrometry with proximity labeling to identify the N-cadherin (CDH2) interactome. We define over 350 proteins in this interactome, nearly 200 of which are unique to CDH2 and not part of the E-cadherin (CDH1) interactome. CDH2-specific interactors comprise primarily adaptor and adhesion proteins that promote junction specialization. Our results provide novel insight into the cardiomyocyte AJ and offer a proteomic atlas for defining the molecular complexes that regulate cardiomyocyte intercellular adhesion. This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Yang Li
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Chelsea D Merkel
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Xuemei Zeng
- Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA 15261, USA
| | - Jonathon A Heier
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Pamela S Cantrell
- Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA 15261, USA
| | - Mai Sun
- Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA 15261, USA
| | - Donna B Stolz
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Nathan A Yates
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA 15261, USA.,University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Adam V Kwiatkowski
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
37
|
Knyazeva A, Krutikov A, Golovkin A, Mishanin A, Pavlov G, Smolina N, Hushkina A, Sejersen T, Sjoberg G, Galagudza M, Kostareva A. Time- and Ventricular-Specific Expression Profiles of Genes Encoding Z-Disk Proteins in Pressure Overload Model of Left Ventricular Hypertrophy. Front Genet 2019; 9:684. [PMID: 30666270 PMCID: PMC6330284 DOI: 10.3389/fgene.2018.00684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/07/2018] [Indexed: 01/20/2023] Open
Abstract
Mechanotransduction is an essential mechanism of transforming external mechanical stimulus to biochemical response. In cardiomyocytes mechanotransduction plays an important role in contraction, stretch sensing and homeostasis regulation. One of the major mechanosensitive area in cardiomyocytes, the Z-disk, consists of numbers of structural and signaling proteins, that may undergo conformational or gene expression changes under pathological stress conditions. In present study we examined a rat model of pressure overload cardiac hypertrophy validated by echocardiographic and histopathological examinations. We revealed, that during hypertrophy progression expression of several genes encoding Z-disk proteins (Actn2, Ldb3, Cmya5, Nebl) is different at early and late points of cardiac remodeling. Moreover, expression patterns of several genes are opposite in myocardium of overloaded left ventricle and hemodynamically unaffected right ventricle, and expression profiles in interventricular septum are more similar to right ventricle. Additionally, we revealed inconsistencies between mRNA and protein level changes of Actn2, one of the major structural Z-disk element. All these findings point out, that investigated Z-disk proteins participate in pathological stress adaptation through undergoing the gene expression changes, and suggest the novel important role of hypertrophic response modulation during different stages of cardiac remodeling.
Collapse
Affiliation(s)
| | | | - Alexey Golovkin
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | | | - Georgii Pavlov
- Saint Petersburg State Academy of Veterinary Medicine, Saint Petersburg, Russia
| | - Natalia Smolina
- Almazov National Medical Research Centre, Saint Petersburg, Russia.,Information Technologies and Programming Faculty, ITMO University, Saint Petersburg, Russia
| | | | - Thomas Sejersen
- Department of Women's and Children's Health, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Gunnar Sjoberg
- Department of Women's and Children's Health, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia.,Information Technologies and Programming Faculty, ITMO University, Saint Petersburg, Russia
| |
Collapse
|
38
|
Chen Z, Maimaiti R, Zhu C, Cai H, Stern A, Mozdziak P, Ge Y, Ford SP, Nathanielsz PW, Guo W. Z-band and M-band titin splicing and regulation by RNA binding motif 20 in striated muscles. J Cell Biochem 2018; 119:9986-9996. [PMID: 30133019 PMCID: PMC6218289 DOI: 10.1002/jcb.27328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/28/2018] [Indexed: 12/12/2022]
Abstract
Titin (TTN) has multifunctional roles in sarcomere assembly, mechanosignaling transduction, and muscle stiffness. TTN splicing generates variable protein sizes with different functions. Therefore, understanding TTN splicing is important to develop a novel treatment for TTN-based diseases. The I-band TTN splicing regulated by RNA binding motif 20 (RBM20) has been extensively studied. However, the Z- and M-band splicing and regulation remain poorly understood. Herein, we aimed to define the Z- and M-band splicing in striated muscles and determined whether RBM20 regulates the Z- and M-band splicing. We discovered four new Z-band TTN splicing variants, and one of them dominates in mouse, rat, sheep, and human hearts. But only one form can be detected in frog and chicken hearts. In skeletal muscles, three new Z repeats (Zr) were detected, and Zr4 to 6 exclusion dominates in the fast muscles, whereas Zr4 skipping dominates in the slow muscle. No developmental changes were detected in the Z-band. In the M-band, two new variants were discovered with alternative 3' splice site in exon363 (Mex5) and alternative 5' splice site in intron 362. However, only the sheep heart expresses two new variants rather than other species. Skeletal muscles express three M-band variants with altered ratios of Mex5 inclusion to Mex5 exclusion. Finally, we revealed that RBM20 does not regulate the Z- and M-band splicing in the heart, but does in skeletal muscles. Taken together, we characterized the Z- and M-band splicing and provided the first evidence of the role of RBM20 in the Z- and M-band TTN splicing.
Collapse
Affiliation(s)
- Zhilong Chen
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi, China
- Department of Animal Science, University of Wyoming, Laramie, Wyoming
| | - Rexiati Maimaiti
- Department of Animal Science, University of Wyoming, Laramie, Wyoming
| | - Chaoqun Zhu
- Department of Animal Science, University of Wyoming, Laramie, Wyoming
| | - Hanfang Cai
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi, China
- Department of Animal Science, University of Wyoming, Laramie, Wyoming
| | - Allysa Stern
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, North Carolina
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, North Carolina
| | - Ying Ge
- Department of Cell and Regenerative Biology, Human Proteomics Program, University of Wisconsin, Madison, Wisconsin
- Department of Chemistry, Human Proteomics Program, University of Wisconsin, Madison, Wisconsin
| | - Stephen P Ford
- Department of Animal Science, University of Wyoming, Laramie, Wyoming
| | | | - Wei Guo
- Department of Animal Science, University of Wyoming, Laramie, Wyoming
| |
Collapse
|
39
|
Wackerhage H, Schoenfeld BJ, Hamilton DL, Lehti M, Hulmi JJ. Stimuli and sensors that initiate skeletal muscle hypertrophy following resistance exercise. J Appl Physiol (1985) 2018; 126:30-43. [PMID: 30335577 DOI: 10.1152/japplphysiol.00685.2018] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
One of the most striking adaptations to exercise is the skeletal muscle hypertrophy that occurs in response to resistance exercise. A large body of work shows that a mammalian target of rapamycin complex 1 (mTORC1)-mediated increase of muscle protein synthesis is the key, but not sole, mechanism by which resistance exercise causes muscle hypertrophy. While much of the hypertrophy signaling cascade has been identified, the initiating, resistance exercise-induced and hypertrophy-stimulating stimuli have remained elusive. For the purpose of this review, we define an initiating, resistance exercise-induced and hypertrophy-stimulating signal as "hypertrophy stimulus," and the sensor of such a signal as "hypertrophy sensor." In this review we discuss our current knowledge of specific mechanical stimuli, damage/injury-associated and metabolic stress-associated triggers, as potential hypertrophy stimuli. Mechanical signals are the prime hypertrophy stimuli candidates, and a filamin-C-BAG3-dependent regulation of mTORC1, Hippo, and autophagy signaling is a plausible albeit still incompletely characterized hypertrophy sensor. Other candidate mechanosensing mechanisms are nuclear deformation-initiated signaling or several mechanisms related to costameres, which are the functional equivalents of focal adhesions in other cells. While exercise-induced muscle damage is probably not essential for hypertrophy, it is still unclear whether and how such muscle damage could augment a hypertrophic response. Interventions that combine blood flow restriction and especially low load resistance exercise suggest that resistance exercise-regulated metabolites could be hypertrophy stimuli, but this is based on indirect evidence and metabolite candidates are poorly characterized.
Collapse
Affiliation(s)
- Henning Wackerhage
- Department of Sport and Exercise Sciences, Technical University of Munich , Munich , Germany
| | | | - D Lee Hamilton
- Faculty of Health, School of Exercise and Nutrition Sciences, Deakin University , Victoria , Australia
| | - Maarit Lehti
- LIKES Research Centre for Physical Activity and Health , Jyväskylä , Finland
| | - Juha J Hulmi
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä , Jyväskylä , Finland
| |
Collapse
|
40
|
Filipova D, Henry M, Rotshteyn T, Brunn A, Carstov M, Deckert M, Hescheler J, Sachinidis A, Pfitzer G, Papadopoulos S. Distinct transcriptomic changes in E14.5 mouse skeletal muscle lacking RYR1 or Cav1.1 converge at E18.5. PLoS One 2018; 13:e0194428. [PMID: 29543863 PMCID: PMC5854361 DOI: 10.1371/journal.pone.0194428] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/04/2018] [Indexed: 12/20/2022] Open
Abstract
In skeletal muscle the coordinated actions of two mechanically coupled Ca2+ channels-the 1,4-dihydropyridine receptor (Cav1.1) and the type 1 ryanodine receptor (RYR1)-underlie the molecular mechanism of rapid cytosolic [Ca2+] increase leading to contraction. While both [Ca2+]i and contractile activity have been implicated in the regulation of myogenesis, less is known about potential specific roles of Cav1.1 and RYR1 in skeletal muscle development. In this study, we analyzed the histology and the transcriptomic changes occurring at E14.5 -the end of primary myogenesis and around the onset of intrauterine limb movement, and at E18.5 -the end of secondary myogenesis, in WT, RYR1-/-, and Cav1.1-/- murine limb skeletal muscle. At E14.5 the muscle histology of both mutants exhibited initial alterations, which became much more severe at E18.5. Immunohistological analysis also revealed higher levels of activated caspase-3 in the Cav1.1-/- muscles at E14.5, indicating an increase in apoptosis. With WT littermates as controls, microarray analyses identified 61 and 97 differentially regulated genes (DEGs) at E14.5, and 493 and 1047 DEGs at E18.5, in RYR1-/- and Cav1.1-/- samples, respectively. Gene enrichment analysis detected no overlap in the affected biological processes and pathways in the two mutants at E14.5, whereas at E18.5 there was a significant overlap of DEGs in both mutants, affecting predominantly processes linked to muscle contraction. Moreover, the E18.5 vs. E14.5 comparison revealed multiple genotype-specific DEGs involved in contraction, cell cycle and miRNA-mediated signaling in WT, neuronal and bone development in RYR1-/-, and lipid metabolism in Cav1.1-/- samples. Taken together, our study reveals discrete changes in the global transcriptome occurring in limb skeletal muscle from E14.5 to E18.5 in WT, RYR1-/- and Cav1.1-/- mice. Our results suggest distinct functional roles for RYR1 and Cav1.1 in skeletal primary and secondary myogenesis.
Collapse
Affiliation(s)
- Dilyana Filipova
- Institute of Vegetative Physiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| | - Margit Henry
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Tamara Rotshteyn
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Anna Brunn
- Department of Neuropathology, University of Cologne, Cologne, Germany
| | - Mariana Carstov
- Department of Neuropathology, University of Cologne, Cologne, Germany
| | - Martina Deckert
- Department of Neuropathology, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Gabriele Pfitzer
- Institute of Vegetative Physiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| | - Symeon Papadopoulos
- Institute of Vegetative Physiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| |
Collapse
|
41
|
Arimura T, Muchir A, Kuwahara M, Morimoto S, Ishikawa T, Du CK, Zhan DY, Nakao S, Machida N, Tanaka R, Yamane Y, Hayashi T, Kimura A. Overexpression of heart-specific small subunit of myosin light chain phosphatase results in heart failure and conduction disturbance. Am J Physiol Heart Circ Physiol 2018; 314:H1192-H1202. [PMID: 29451818 DOI: 10.1152/ajpheart.00696.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mutations in genes encoding components of the sarcomere cause cardiomyopathy, which is often associated with abnormal Ca2+ sensitivity of muscle contraction. We have previously shown that a heart-specific myosin light chain phosphatase small subunit (hHS-M21) increases the Ca2+ sensitivity of muscle contraction. The aim of the present study was to investigate the function of hHS-M21 in vivo and the causative role of abnormal Ca2+ sensitivity in cardiomyopathy. We generated transgenic mice with cardiac-specific overexpression of hHS-M21. We confirmed that hHS-M21 increased the Ca2+ sensitivity of cardiac muscle contraction in vivo, which was not followed by an increased phosphorylation of myosin light chain 2 isoforms. hHS-M21 transgenic mice developed severe systolic dysfunction with myocardial fibrosis and degeneration of cardiomyocytes in association with sinus bradycardia and atrioventricular conduction defect. The contractile dysfunction and cardiac fibrosis were improved by treatment with the Rho kinase inhibitor fasudil. Our findings suggested that the overexpression of hHS-M21 results in cardiac dysfunction and conduction disturbance via non-myosin light chain 2 phosphorylation-dependent regulation. NEW & NOTEWORTHY The present study is the first to develop mice with transgenic overexpression of a heart-specific myosin light chain phosphatase small subunit (hHS-M21) and to examine the effects of hHS-M21 on cardiac function. Elevation of hHS-M21 induced heart failure with myocardial fibrosis and degeneration of cardiomyocytes accompanied by supraventricular arrhythmias.
Collapse
Affiliation(s)
- Takuro Arimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University , Tokyo , Japan
| | - Antoine Muchir
- Department of Medicine and Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University , Columbia, New York
| | - Masayoshi Kuwahara
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agriculture and Life Sciences, The University of Tokyo , Tokyo , Japan
| | - Sachio Morimoto
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University , Fukuoka , Japan
| | - Taisuke Ishikawa
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University , Tokyo , Japan
| | - Cheng-Kun Du
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University , Fukuoka , Japan
| | - Dong-Yun Zhan
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University , Fukuoka , Japan
| | - Shu Nakao
- Department of Veterinary Clinical Oncology, Faculty of Agriculture, Tokyo University of Agriculture and Technology , Tokyo , Japan
| | - Noboru Machida
- Department of Veterinary Clinical Oncology, Faculty of Agriculture, Tokyo University of Agriculture and Technology , Tokyo , Japan
| | - Ryo Tanaka
- Department of Veterinary Surgery, Faculty of Agriculture, Tokyo University of Agriculture and Technology , Tokyo , Japan
| | - Yoshihisa Yamane
- Department of Veterinary Surgery, Faculty of Agriculture, Tokyo University of Agriculture and Technology , Tokyo , Japan
| | - Takeharu Hayashi
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University , Tokyo , Japan
| | - Akinori Kimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University , Tokyo , Japan
| |
Collapse
|
42
|
Sato Y, Ohtsubo H, Nihei N, Kaneko T, Sato Y, Adachi SI, Kondo S, Nakamura M, Mizunoya W, Iida H, Tatsumi R, Rada C, Yoshizawa F. Apobec2 deficiency causes mitochondrial defects and mitophagy in skeletal muscle. FASEB J 2018; 32:1428-1439. [PMID: 29127187 PMCID: PMC5892721 DOI: 10.1096/fj.201700493r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Apobec2 is a member of the activation-induced deaminase/apolipoprotein B mRNA editing enzyme catalytic polypeptide cytidine deaminase family expressed in differentiated skeletal and cardiac muscle. We previously reported that Apobec2 deficiency in mice leads to a shift in muscle fiber type, myopathy, and diminished muscle mass. However, the mechanisms of myopathy caused by Apobec2 deficiency and its physiologic functions are unclear. Here we show that, although Apobec2 localizes to the sarcomeric Z-lines in mouse tissue and cultured myotubes, the sarcomeric structure is not affected in Apobec2-deficient muscle. In contrast, electron microscopy reveals enlarged mitochondria and mitochondria engulfed by autophagic vacuoles, suggesting that Apobec2 deficiency causes mitochondrial defects leading to increased mitophagy in skeletal muscle. Indeed, Apobec2 deficiency results in increased reactive oxygen species generation and depolarized mitochondria, leading to mitophagy as a defensive response. Furthermore, the exercise capacity of Apobec2-/- mice is impaired, implying Apobec2 deficiency results in ongoing muscle dysfunction. The presence of rimmed vacuoles in myofibers from 10-mo-old mice suggests that the chronic muscle damage impairs normal autophagy. We conclude that Apobec2 deficiency causes mitochondrial defects that increase muscle mitophagy, leading to myopathy and atrophy. Our findings demonstrate that Apobec2 is required for mitochondrial homeostasis to maintain normal skeletal muscle function.-Sato, Y., Ohtsubo, H., Nihei, N., Kaneko, T., Sato, Y., Adachi, S.-I., Kondo, S., Nakamura, M., Mizunoya, W., Iida, H., Tatsumi, R., Rada, C., Yoshizawa, F. Apobec2 deficiency causes mitochondrial defects and mitophagy in skeletal muscle.
Collapse
Affiliation(s)
- Yusuke Sato
- Department of Agrobiology and Bioresources, Utsunomiya University, Tochigi, Japan
| | - Hideaki Ohtsubo
- Department of Animal and Marine Bioresource Sciences, Kyushu University, Fukuoka, Japan
| | - Naohiro Nihei
- Department of Agrobiology and Bioresources, Utsunomiya University, Tochigi, Japan
| | - Takane Kaneko
- Department of Animal and Marine Bioresource Sciences, Kyushu University, Fukuoka, Japan
| | - Yoriko Sato
- United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Shin-Ichi Adachi
- Department of Agrobiology and Bioresources, Utsunomiya University, Tochigi, Japan
| | - Shinji Kondo
- Department of Agrobiology and Bioresources, Utsunomiya University, Tochigi, Japan
| | - Mako Nakamura
- Department of Animal and Marine Bioresource Sciences, Kyushu University, Fukuoka, Japan
| | - Wataru Mizunoya
- Department of Animal and Marine Bioresource Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroshi Iida
- Department of Animal and Marine Bioresource Sciences, Kyushu University, Fukuoka, Japan
| | - Ryuichi Tatsumi
- Department of Animal and Marine Bioresource Sciences, Kyushu University, Fukuoka, Japan
| | - Cristina Rada
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Fumiaki Yoshizawa
- Department of Agrobiology and Bioresources, Utsunomiya University, Tochigi, Japan.,United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
43
|
Ye M, Ye F, He L, Luo B, Yang F, Cui C, Zhao X, Yin H, Li D, Xu H, Wang Y, Zhu Q. Transcriptomic analysis of chicken Myozenin 3 regulation reveals its potential role in cell proliferation. PLoS One 2017; 12:e0189476. [PMID: 29236749 PMCID: PMC5728575 DOI: 10.1371/journal.pone.0189476] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 11/28/2017] [Indexed: 11/19/2022] Open
Abstract
Embryonic muscle development and fibre type differentiation has always been a topic of great importance due to its impact on both human health and farm animal financial values. Myozenin3 (Myoz3) is an important candidate gene that may regulate these processes. In the current study, we knocked down and overexpressed Myoz3 in chicken embryonic fibroblasts (CEFs) and chicken myoblasts, then utilized RNA-seq technology to screen genes, pathways and biological processes associated with Myoz3. Multiple differentially expressed genes were identified, including MYH10, MYLK2, NFAM1, MYL4, MYL9, PDZLIM1; those can in turn regulate each other and influence the development of muscle fibres. Gene ontology (GO) terms including some involved in positive regulation of cell proliferation were enriched. We further validated our results by testing the activity of cells by cell counting kit-8(CCK-8) and confirmed that under the condition of Myoz3 overexpression, the proliferation rate of CEFs and myoblasts was significantly upregulated, in addition, expression level of fast muscle specific gene was also significantly upregulated in myoblasts. Pathway enrichment analysis revealed that the PPAR (Peroxisome Proliferator-Activated Receptor) pathway was enriched, suggesting the possibility that Myoz3 regulates muscle fibre development and differentiation through the PPAR pathway. Our results provide valuable evidence regarding the regulatory functions of Myoz3 in embryonic cells by screening multiple candidate genes, biological processes and pathways associated with Myoz3.
Collapse
Affiliation(s)
- Maosen Ye
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Fei Ye
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Liutao He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Bin Luo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Fuling Yang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Can Cui
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Xiaoling Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Huadong Yin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Diyan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Hengyong Xu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
- * E-mail: (YW); (QZ)
| | - Qing Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
- * E-mail: (YW); (QZ)
| |
Collapse
|
44
|
Downregulation of Profilin-1 Expression Attenuates Cardiomyocytes Hypertrophy and Apoptosis Induced by Advanced Glycation End Products in H9c2 Cells. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9716087. [PMID: 29238726 PMCID: PMC5697376 DOI: 10.1155/2017/9716087] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 09/08/2017] [Accepted: 10/04/2017] [Indexed: 12/21/2022]
Abstract
Cardiomyocytes hypertrophy and apoptosis induced by advanced glycation end products (AGEs) is the crucial pathological foundation contributing to the onset and development of diabetic cardiomyopathy (DCM). However, the mechanism remains poorly understood. Here, we report that profilin-1 (PFN-1), a well-known actin-binding protein, serves as a potent regulator in AGEs-induced cardiomyocytes hypertrophy and apoptosis. PFN-1 was upregulated in AGEs-treated H9c2 cells, which was associated with increased cardiomyocytes hypertrophy and apoptosis. Silencing PFN-1 expression remarkably attenuated AGEs-induced H9c2 cell hypertrophy and apoptosis. Mechanistically, AGEs increased PFN-1 expression through elevating ROS production and RhoA and ROCK2 expression. Consequently, elevated PFN-1 promoted actin cytoskeleton disorganization. When either ROS production/ROCK activation was blocked or cells were treated with Cytochalasin D (actin depolymerizer), H9c2 cells were protected against AGEs-induced cardiac myocyte abnormalities, concomitantly with downregulated expression of PFN-1 and improved actin cytoskeleton alteration. Collectively, these data suggest that PFN-1 may play an important role in AGEs-induced hypertrophy and apoptosis in H9c2 cells.
Collapse
|
45
|
Potts GK, McNally RM, Blanco R, You J, Hebert AS, Westphall MS, Coon JJ, Hornberger TA. A map of the phosphoproteomic alterations that occur after a bout of maximal-intensity contractions. J Physiol 2017; 595:5209-5226. [PMID: 28542873 PMCID: PMC5538225 DOI: 10.1113/jp273904] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 05/11/2017] [Indexed: 12/25/2022] Open
Abstract
KEY POINTS Mechanical signals play a critical role in the regulation of muscle mass, but the molecules that sense mechanical signals and convert this stimulus into the biochemical events that regulate muscle mass remain ill-defined. Here we report a mass spectrometry-based workflow to study the changes in protein phosphorylation that occur in mouse skeletal muscle 1 h after a bout of electrically evoked maximal-intensity contractions (MICs). Our dataset provides the first comprehensive map of the MIC-regulated phosphoproteome. Using unbiased bioinformatics approaches, we demonstrate that our dataset leads to the identification of many well-known MIC-regulated signalling pathways, as well as to a plethora of novel MIC-regulated events. We expect that our dataset will serve as a fundamentally important resource for muscle biologists, and help to lay the foundation for entirely new hypotheses in the field. ABSTRACT The maintenance of skeletal muscle mass is essential for health and quality of life. It is well recognized that maximal-intensity contractions, such as those which occur during resistance exercise, promote an increase in muscle mass. Yet, the molecules that sense the mechanical information and convert it into the signalling events (e.g. phosphorylation) that drive the increase in muscle mass remain undefined. Here we describe a phosphoproteomics workflow to examine the effects of electrically evoked maximal-intensity contractions (MICs) on protein phosphorylation in mouse skeletal muscle. While a preliminary phosphoproteomics experiment successfully identified a number of MIC-regulated phosphorylation events, a large proportion of these identifications were present on highly abundant myofibrillar proteins. We subsequently incorporated a centrifugation-based fractionation step to deplete the highly abundant myofibrillar proteins and performed a second phosphoproteomics experiment. In total, we identified 5983 unique phosphorylation sites of which 663 were found to be regulated by MIC. GO term enrichment, phosphorylation motif analyses, and kinase-substrate predictions indicated that the MIC-regulated phosphorylation sites were chiefly modified by mTOR, as well as multiple isoforms of the MAPKs and CAMKs. Moreover, a high proportion of the regulated phosphorylation sites were found on proteins that are associated with the Z-disc, with over 74% of the Z-disc proteins experiencing robust changes in phosphorylation. Finally, our analyses revealed that the phosphorylation state of two Z-disc kinases (striated muscle-specific serine/threonine protein kinase and obscurin) was dramatically altered by MIC, and we propose ways these kinases could play a fundamental role in skeletal muscle mechanotransduction.
Collapse
Affiliation(s)
- Gregory K. Potts
- Department of ChemistryUniversity of Wisconsin – MadisonMadisonWIUSA
- Genome Center of WisconsinUniversity of Wisconsin – MadisonMadisonWIUSA
| | - Rachel M. McNally
- Department of Comparative BiosciencesUniversity of Wisconsin – MadisonMadisonWIUSA
- School of Veterinary MedicineUniversity of Wisconsin – MadisonMadisonWIUSA
| | - Rocky Blanco
- Department of Comparative BiosciencesUniversity of Wisconsin – MadisonMadisonWIUSA
- School of Veterinary MedicineUniversity of Wisconsin – MadisonMadisonWIUSA
| | - Jae‐Sung You
- Department of Comparative BiosciencesUniversity of Wisconsin – MadisonMadisonWIUSA
- School of Veterinary MedicineUniversity of Wisconsin – MadisonMadisonWIUSA
| | - Alexander S. Hebert
- Genome Center of WisconsinUniversity of Wisconsin – MadisonMadisonWIUSA
- Department of Biomolecular ChemistryUniversity of Wisconsin – MadisonMadisonWIUSA
| | | | - Joshua J. Coon
- Department of ChemistryUniversity of Wisconsin – MadisonMadisonWIUSA
- Genome Center of WisconsinUniversity of Wisconsin – MadisonMadisonWIUSA
- Department of Biomolecular ChemistryUniversity of Wisconsin – MadisonMadisonWIUSA
| | - Troy A. Hornberger
- Department of Comparative BiosciencesUniversity of Wisconsin – MadisonMadisonWIUSA
- School of Veterinary MedicineUniversity of Wisconsin – MadisonMadisonWIUSA
| |
Collapse
|
46
|
Ryanodine receptors are part of the myospryn complex in cardiac muscle. Sci Rep 2017; 7:6312. [PMID: 28740084 PMCID: PMC5524797 DOI: 10.1038/s41598-017-06395-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/12/2017] [Indexed: 01/28/2023] Open
Abstract
The Cardiomyopathy-associated gene 5 (Cmya5) encodes myospryn, a large tripartite motif (TRIM)-related protein found predominantly in cardiac and skeletal muscle. Cmya5 is an expression biomarker for a number of diseases affecting striated muscle and may also be a schizophrenia risk gene. To further understand the function of myospryn in striated muscle, we searched for additional myospryn paralogs. Here we identify a novel muscle-expressed TRIM-related protein minispryn, encoded by Fsd2, that has extensive sequence similarity with the C-terminus of myospryn. Cmya5 and Fsd2 appear to have originated by a chromosomal duplication and are found within evolutionarily-conserved gene clusters on different chromosomes. Using immunoaffinity purification and mass spectrometry we show that minispryn co-purifies with myospryn and the major cardiac ryanodine receptor (RyR2) from heart. Accordingly, myospryn, minispryn and RyR2 co-localise at the junctional sarcoplasmic reticulum of isolated cardiomyocytes. Myospryn redistributes RyR2 into clusters when co-expressed in heterologous cells whereas minispryn lacks this activity. Together these data suggest a novel role for the myospryn complex in the assembly of ryanodine receptor clusters in striated muscle.
Collapse
|
47
|
Rusu M, Hu Z, Taylor KA, Trinick J. Structure of isolated Z-disks from honeybee flight muscle. J Muscle Res Cell Motil 2017; 38:241-250. [PMID: 28733815 PMCID: PMC5660141 DOI: 10.1007/s10974-017-9477-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 07/14/2017] [Indexed: 12/11/2022]
Abstract
The Z-disk is a complex structure comprising some 40 proteins that are involved in the transmission of force developed during muscle contraction and in important signalling pathways that govern muscle homeostasis. In the Z-disk the ends of antiparallel thin filaments from adjacent sarcomeres are crosslinked by α-actinin. The structure of the Z-disk lattice varies greatly throughout the animal kingdom. In vertebrates the thin filaments form a tetragonal lattice, whereas invertebrate flight muscle has a hexagonal lattice. The width of the Z-disk varies considerably and correlates with the number of α-actinin bridges. A detailed description at a high resolution of the Z-disk lattice is needed in order to better understand muscle function and disease. The molecular architecture of the Z-disk lattice in honeybee (Apis mellifera) is known from plastic embedded thin sections to a resolution of 7 nm, which is not sufficient to dock component protein crystal structures. It has been shown that sectioning is a damaging process that leads to the loss of finer details present in biological specimens. However, the Apis Z-disk is a thin structure (120 nm) suitable for cryo EM. We have isolated intact honeybee Z-disks from indirect flight muscle, thus obviating the need of plastic sectioning. We have employed cryo electron tomography and image processing to investigate the arrangement of proteins within the hexagonal lattice of the Apis Z-disk. The resolution obtained, ~6 nm, was probably limited by damage caused by the harshness of the conditions used to extract the myofibrils and isolate the Z-disks.
Collapse
Affiliation(s)
- Mara Rusu
- Astbury Center, University of Leeds, Leeds, LS2 9JT, UK
| | - Zhongjun Hu
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32306-4380, USA
| | - Kenneth A Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32306-4380, USA
| | - John Trinick
- Astbury Center, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
48
|
Dierck F, Kuhn C, Rohr C, Hille S, Braune J, Sossalla S, Molt S, van der Ven PFM, Fürst DO, Frey N. The novel cardiac z-disc protein CEFIP regulates cardiomyocyte hypertrophy by modulating calcineurin signaling. J Biol Chem 2017; 292:15180-15191. [PMID: 28717008 DOI: 10.1074/jbc.m117.786764] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/04/2017] [Indexed: 11/06/2022] Open
Abstract
The z-disc is a structural component at the lateral borders of the sarcomere and is important for mechanical stability and contractility of both cardiac and skeletal muscles. Of note, the sarcomeric z-disc also represents a nodal point in cardiomyocyte function and signaling. Mutations of numerous z-disc proteins are associated with cardiomyopathies and muscle diseases. To identify additional z-disc proteins that might contribute to cardiac disease, we employed an in silico screen for cardiac-enriched cDNAs. This screen yielded a previously uncharacterized protein named cardiac-enriched FHL2-interacting protein (CEFIP), which exhibited a heart- and skeletal muscle-specific expression profile. Importantly, CEFIP was located at the z-disc and was up-regulated in several models of cardiomyopathy. We also found that CEFIP overexpression induced the fetal gene program and cardiomyocyte hypertrophy. Yeast two-hybrid screens revealed that CEFIP interacts with the calcineurin-binding protein four and a half LIM domains 2 (FHL2). Because FHL2 binds calcineurin, a phosphatase controlling hypertrophic signaling, we examined the effects of CEFIP on the calcineurin/nuclear factor of activated T-cell (NFAT) pathway. These experiments revealed that CEFIP overexpression further enhances calcineurin-dependent hypertrophic signal transduction, and its knockdown repressed hypertrophy and calcineurin/NFAT activity. In summary, we report on a previously uncharacterized protein CEFIP that modulates calcineurin/NFAT signaling in cardiomyocytes, a finding with possible implications for the pathogenesis of cardiomyopathy.
Collapse
Affiliation(s)
- Franziska Dierck
- From the Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, 24105 Kiel.,the DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105 Kiel
| | - Christian Kuhn
- From the Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, 24105 Kiel.,the DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105 Kiel
| | - Claudia Rohr
- the Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, and
| | - Susanne Hille
- From the Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, 24105 Kiel.,the DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105 Kiel
| | - Julia Braune
- the Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Samuel Sossalla
- From the Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, 24105 Kiel.,the DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105 Kiel
| | - Sibylle Molt
- the Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Peter F M van der Ven
- the Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Dieter O Fürst
- the Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Norbert Frey
- From the Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, 24105 Kiel, .,the DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105 Kiel
| |
Collapse
|
49
|
Higashi H, Ogimoto A, Inoue K, Tokunaga N, Iio C, Aono J, Yoshida S, Izutani H, Tabara Y, Okura T, Higaki J, Ikeda S. Eccentric Left Ventricular Hypertrophy in Aortic Stenosis Caused by Unicuspid Aortic Valve. Circ J 2017; 81:895-897. [PMID: 28123168 DOI: 10.1253/circj.cj-16-0945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Haruhiko Higashi
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University Graduate School of Medicine
| | - Akiyoshi Ogimoto
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University Graduate School of Medicine
| | - Katsuji Inoue
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University Graduate School of Medicine
| | - Naohito Tokunaga
- Division of Analytical Bio-Medicine, Advanced Research Support Center, Ehime University
| | - Chiharuko Iio
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University Graduate School of Medicine
| | - Jun Aono
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University Graduate School of Medicine
| | - Satoshi Yoshida
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University Graduate School of Medicine
| | - Hironori Izutani
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine
| | - Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine
| | - Takafumi Okura
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University Graduate School of Medicine
| | - Jitsuo Higaki
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University Graduate School of Medicine
| | - Shuntaro Ikeda
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University Graduate School of Medicine
| |
Collapse
|
50
|
Hempel A, Kühl SJ, Rothe M, Rao Tata P, Sirbu IO, Vainio SJ, Kühl M. The CapZ interacting protein Rcsd1 is required for cardiogenesis downstream of Wnt11a in Xenopus laevis. Dev Biol 2017; 424:28-39. [PMID: 28237811 DOI: 10.1016/j.ydbio.2017.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/20/2017] [Accepted: 02/20/2017] [Indexed: 11/17/2022]
Abstract
Wnt proteins are critical for embryonic cardiogenesis and cardiomyogenesis by regulating different intracellular signalling pathways. Whereas canonical Wnt/β-catenin signalling is required for mesoderm induction and proliferation of cardiac progenitor cells, β-catenin independent, non-canonical Wnt signalling regulates cardiac specification and terminal differentiation. Although the diverse cardiac malformations associated with the loss of non-canonical Wnt11 in mice such as outflow tract (OFT) defects, reduced ventricular trabeculation, myofibrillar disorganization and reduced cardiac marker gene expression are well described, the underlying molecular mechanisms are still not completely understood. Here we aimed to further characterize Wnt11 mediated signal transduction during vertebrate cardiogenesis. Using Xenopus as a model system, we show by loss of function and corresponding rescue experiments that the non-canonical Wnt signalling mediator Rcsd1 is required downstream of Wnt11 for ventricular trabeculation, terminal differentiation of cardiomyocytes and cardiac morphogenesis. We here place Rcsd1 downstream of Wnt11 during cardiac development thereby providing a novel mechanism for how non-canonical Wnt signalling regulates vertebrate cardiogenesis.
Collapse
Affiliation(s)
- Annemarie Hempel
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, D-89081 Ulm, Germany; International Graduate School in Molecular Medicine Ulm, IGradU, Ulm University, 89069 Ulm, Germany
| | - Susanne J Kühl
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, D-89081 Ulm, Germany
| | - Melanie Rothe
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, D-89081 Ulm, Germany; International Graduate School in Molecular Medicine Ulm, IGradU, Ulm University, 89069 Ulm, Germany
| | - Purushothama Rao Tata
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, D-89081 Ulm, Germany; International Graduate School in Molecular Medicine Ulm, IGradU, Ulm University, 89069 Ulm, Germany
| | - Ioan Ovidiu Sirbu
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, D-89081 Ulm, Germany
| | - Seppo J Vainio
- Faculty of Biochemistry and Molecular Medicine, BioCenter Oulu and InfoTech Oulu,University of Oulu, Aapistie 5, FIN-90014, Finland
| | - Michael Kühl
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, D-89081 Ulm, Germany.
| |
Collapse
|