1
|
Xu X, Wang X, Li Y, Chen R, Wen H, Wang Y, Ma G. Research progress of ankyrin repeat domain 1 protein: an updated review. Cell Mol Biol Lett 2024; 29:131. [PMID: 39420247 PMCID: PMC11488291 DOI: 10.1186/s11658-024-00647-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Ankyrin repeat domain 1 (Ankrd1) is an acute response protein that belongs to the muscle ankyrin repeat protein (MARP) family. Accumulating evidence has revealed that Ankrd1 plays a crucial role in a wide range of biological processes and diseases. This review consolidates current knowledge on Ankrd1's functions in myocardium and skeletal muscle development, neurogenesis, cancer, bone formation, angiogenesis, wound healing, fibrosis, apoptosis, inflammation, and infection. The comprehensive profile of Ankrd1 in cardiovascular diseases, myopathy, and its potential as a candidate prognostic and diagnostic biomarker are also discussed. In the future, more studies of Ankrd1 are warranted to clarify its role in diseases and assess its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xusan Xu
- Maternal and Child Research Institute, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China
| | - Xiaoxia Wang
- Department of Neurology, Longjiang Hospital, Foshan, 528300, China
| | - Yu Li
- Department of Pediatrics, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China
| | - Riling Chen
- Department of Pediatrics, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China
| | - Houlang Wen
- Medical Genetics Laboratory, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China.
| | - Yajun Wang
- Respiratory Research Institute, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China.
| | - Guoda Ma
- Maternal and Child Research Institute, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China.
| |
Collapse
|
2
|
Jurida L, Werner S, Knapp F, Niemann B, Li L, Grün D, Wirth S, Weber A, Beuerlein K, Liebetrau C, Wiedenroth CB, Guth S, Kojonazarov B, Jafari L, Weissmann N, Günther S, Braun T, Bartkuhn M, Schermuly RT, Dorfmüller P, Yin X, Mayr M, Schmitz ML, Czech L, Schlüter KD, Schulz R, Rohrbach S, Kracht M. A common gene signature of the right ventricle in failing rat and human hearts. NATURE CARDIOVASCULAR RESEARCH 2024; 3:819-840. [PMID: 39196177 PMCID: PMC11358011 DOI: 10.1038/s44161-024-00485-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 05/02/2024] [Indexed: 08/29/2024]
Abstract
The molecular mechanisms of progressive right heart failure are incompletely understood. In this study, we systematically examined transcriptomic changes occurring over months in isolated cardiomyocytes or whole heart tissues from failing right and left ventricles in rat models of pulmonary artery banding (PAB) or aortic banding (AOB). Detailed bioinformatics analyses resulted in the identification of gene signature, protein and transcription factor networks specific to ventricles and compensated or decompensated disease states. Proteomic and RNA-FISH analyses confirmed PAB-mediated regulation of key genes and revealed spatially heterogeneous mRNA expression in the heart. Intersection of rat PAB-specific gene sets with transcriptome datasets from human patients with chronic thromboembolic pulmonary hypertension (CTEPH) led to the identification of more than 50 genes whose expression levels correlated with the severity of right heart disease, including multiple matrix-regulating and secreted factors. These data define a conserved, differentially regulated genetic network associated with right heart failure in rats and humans.
Collapse
Affiliation(s)
- Liane Jurida
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Sebastian Werner
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Fabienne Knapp
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | - Bernd Niemann
- Department of Cardiac and Vascular Surgery, Justus Liebig University, Giessen, Germany
| | - Ling Li
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | - Dimitri Grün
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Stefanie Wirth
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Axel Weber
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Knut Beuerlein
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Christoph Liebetrau
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | | | - Stefan Guth
- Department of Thoracic Surgery, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Baktybek Kojonazarov
- Institute for Lung Health, Justus Liebig University, Giessen, Germany
- Medical Clinic II, Justus Liebig University, Giessen, Germany
- Cardio-Pulmonary Institute, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Leili Jafari
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Norbert Weissmann
- Medical Clinic II, Justus Liebig University, Giessen, Germany
- Cardio-Pulmonary Institute, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Günther
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Braun
- Cardio-Pulmonary Institute, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Marek Bartkuhn
- Cardio-Pulmonary Institute, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Science Unit for Basic and Clinical Medicine, Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
| | - Ralph T Schermuly
- Medical Clinic II, Justus Liebig University, Giessen, Germany
- Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Peter Dorfmüller
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- Department of Internal Medicine, Justus Liebig University Giessen, Giessen, Germany
- Institute of Pathology, Justus Liebig University Giessen, Giessen, Germany
| | - Xiaoke Yin
- National Heart and Lung Institute, Faculty of Medicine,Imperial College London, London, UK
| | - Manuel Mayr
- National Heart and Lung Institute, Faculty of Medicine,Imperial College London, London, UK
| | - M Lienhard Schmitz
- German Center for Lung Research (DZL), Giessen, Germany
- Institute of Biochemistry, Justus Liebig University, Giessen, Germany
| | - Laureen Czech
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | | | - Rainer Schulz
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | - Susanne Rohrbach
- Department of Physiology, Justus Liebig University, Giessen, Germany.
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany.
- Cardio-Pulmonary Institute, Giessen, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany.
- German Center for Lung Research (DZL), Giessen, Germany.
| |
Collapse
|
3
|
Xie R, Yuan S, Hu G, Zhan J, Jin K, Tang Y, Fan J, Zhao Y, Wang F, Chen C, Wang DW, Li H. Nuclear AGO2 promotes myocardial remodeling by activating ANKRD1 transcription in failing hearts. Mol Ther 2024; 32:1578-1594. [PMID: 38475992 PMCID: PMC11081878 DOI: 10.1016/j.ymthe.2024.03.018] [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: 10/13/2023] [Revised: 02/01/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024] Open
Abstract
Heart failure (HF) is manifested by transcriptional and posttranscriptional reprogramming of critical genes. Multiple studies have revealed that microRNAs could translocate into subcellular organelles such as the nucleus to modify gene expression. However, the functional property of subcellular Argonaute2 (AGO2), the core member of the microRNA machinery, has remained elusive in HF. AGO2 was found to be localized in both the cytoplasm and nucleus of cardiomyocytes, and robustly increased in the failing hearts of patients and animal models. We demonstrated that nuclear AGO2 rather than cytosolic AGO2 overexpression by recombinant adeno-associated virus (serotype 9) with cardiomyocyte-specific troponin T promoter exacerbated the cardiac dysfunction in transverse aortic constriction (TAC)-operated mice. Mechanistically, nuclear AGO2 activates the transcription of ANKRD1, encoding ankyrin repeat domain-containing protein 1 (ANKRD1), which also has a dual function in the cytoplasm as part of the I-band of the sarcomere and in the nucleus as a transcriptional cofactor. Overexpression of nuclear ANKRD1 recaptured some key features of cardiac remodeling by inducing pathological MYH7 activation, whereas cytosolic ANKRD1 seemed cardioprotective. For clinical practice, we found ivermectin, an antiparasite drug, and ANPep, an ANKRD1 nuclear location signal mimetic peptide, were able to prevent ANKRD1 nuclear import, resulting in the improvement of cardiac performance in TAC-induced HF.
Collapse
Affiliation(s)
- Rong Xie
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Shuai Yuan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Guo Hu
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Jiabing Zhan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Kunying Jin
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Yuyan Tang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Jiahui Fan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Yanru Zhao
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Feng Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| |
Collapse
|
4
|
Jolfayi AG, Kohansal E, Ghasemi S, Naderi N, Hesami M, MozafaryBazargany M, Moghadam MH, Fazelifar AF, Maleki M, Kalayinia S. Exploring TTN variants as genetic insights into cardiomyopathy pathogenesis and potential emerging clues to molecular mechanisms in cardiomyopathies. Sci Rep 2024; 14:5313. [PMID: 38438525 PMCID: PMC10912352 DOI: 10.1038/s41598-024-56154-7] [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: 11/22/2023] [Accepted: 03/01/2024] [Indexed: 03/06/2024] Open
Abstract
The giant protein titin (TTN) is a sarcomeric protein that forms the myofibrillar backbone for the components of the contractile machinery which plays a crucial role in muscle disorders and cardiomyopathies. Diagnosing TTN pathogenic variants has important implications for patient management and genetic counseling. Genetic testing for TTN variants can help identify individuals at risk for developing cardiomyopathies, allowing for early intervention and personalized treatment strategies. Furthermore, identifying TTN variants can inform prognosis and guide therapeutic decisions. Deciphering the intricate genotype-phenotype correlations between TTN variants and their pathologic traits in cardiomyopathies is imperative for gene-based diagnosis, risk assessment, and personalized clinical management. With the increasing use of next-generation sequencing (NGS), a high number of variants in the TTN gene have been detected in patients with cardiomyopathies. However, not all TTN variants detected in cardiomyopathy cohorts can be assumed to be disease-causing. The interpretation of TTN variants remains challenging due to high background population variation. This narrative review aimed to comprehensively summarize current evidence on TTN variants identified in published cardiomyopathy studies and determine which specific variants are likely pathogenic contributors to cardiomyopathy development.
Collapse
Affiliation(s)
- Amir Ghaffari Jolfayi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Erfan Kohansal
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Serwa Ghasemi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Naderi
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahshid Hesami
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Hosseini Moghadam
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Farjam Fazelifar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Kalayinia
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Orgil BO, Purevjav E. Molecular Pathways and Animal Models of Cardiomyopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:991-1019. [PMID: 38884766 DOI: 10.1007/978-3-031-44087-8_64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Cardiomyopathies are a heterogeneous group of disorders of the heart muscle that ultimately result in congestive heart failure. Rapid progress in genetics, molecular and cellular biology with breakthrough innovative genetic-engineering techniques, such as next-generation sequencing and multiomics platforms, stem cell reprogramming, as well as novel groundbreaking gene-editing systems over the past 25 years has greatly improved the understanding of pathogenic signaling pathways in inherited cardiomyopathies. This chapter will focus on intracellular and intercellular molecular signaling pathways that are activated by a genetic insult in cardiomyocytes to maintain tissue and organ level regulation and resultant cardiac remodeling in certain forms of cardiomyopathies. In addition, animal models of different clinical forms of human cardiomyopathies with their summaries of triggered key molecules and signaling pathways will be described.
Collapse
Affiliation(s)
- Buyan-Ochir Orgil
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
6
|
Poelmann RE, Jongbloed MRM, DeRuiter MC. Total Anomalous Pulmonary Venous Connections, Human Genetics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:593-598. [PMID: 38884735 DOI: 10.1007/978-3-031-44087-8_33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Partial anomalous pulmonary venous connections (PAVC) have been found after abnormal gene expressions involving several syndromes. Total anomalous pulmonary venous connection (TAPVC) is found in conjunction with heterotaxia syndrome as well as several other syndromes. It has been reported with an autosomal dominance with variable expression and incomplete penetrance. The occurrence is also related to environmental factors which may superimpose on a familial susceptibility for TAPVC. Many pathways are involved in the normal development of the pulmonary venous connections and as a consequence disturbance of many genetic and epigenetic pathways lead to partial or total pulmonary venous misconnections. In this chapter, an overview of current knowledge regarding human genetics of anomalous venous connections is provided.
Collapse
Affiliation(s)
- R E Poelmann
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - M R M Jongbloed
- Department Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - M C DeRuiter
- Department Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
7
|
Rinkūnaitė I, Šimoliūnas E, Alksnė M, Bartkutė G, Labeit S, Bukelskienė V, Bogomolovas J. Genetic Ablation of Ankrd1 Mitigates Cardiac Damage during Experimental Autoimmune Myocarditis in Mice. Biomolecules 2022; 12:biom12121898. [PMID: 36551326 PMCID: PMC9775225 DOI: 10.3390/biom12121898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/05/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Myocarditis (MC) is an inflammatory disease of the myocardium that can cause sudden death in the acute phase, and dilated cardiomyopathy (DCM) with chronic heart failure as its major long-term outcome. However, the molecular mechanisms beyond the acute MC phase remain poorly understood. The ankyrin repeat domain 1 (ANKRD1) is a functionally pleiotropic stress/stretch-inducible protein, which can modulate cardiac stress response during various forms of pathological stimuli; however, its involvement in post-MC cardiac remodeling leading to DCM is not known. To address this, we induced experimental autoimmune myocarditis (EAM) in ANKRD1-deficient mice, and evaluated post-MC consequences at the DCM stage mice hearts. We demonstrated that ANKRD1 does not significantly modulate heart failure; nevertheless, the genetic ablation of Ankrd1 blunted the cardiac damage/remodeling and preserved heart function during post-MC DCM.
Collapse
Affiliation(s)
- Ieva Rinkūnaitė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Egidijus Šimoliūnas
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Milda Alksnė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Gabrielė Bartkutė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Siegfried Labeit
- DZHK Partner Site Mannheim-Heidelberg, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Myomedix GmbH, 69151 Neckargemünd, Germany
| | - Virginija Bukelskienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Julius Bogomolovas
- Department of Medicine, School of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
- Correspondence:
| |
Collapse
|
8
|
Hindmarch CCT, Tian L, Xiong PY, Potus F, Bentley RET, Al-Qazazi R, Prins KW, Archer SL. An integrated proteomic and transcriptomic signature of the failing right ventricle in monocrotaline induced pulmonary arterial hypertension in male rats. Front Physiol 2022; 13:966454. [PMID: 36388115 PMCID: PMC9664166 DOI: 10.3389/fphys.2022.966454] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/19/2022] [Indexed: 01/25/2023] Open
Abstract
Aim: Pulmonary arterial hypertension (PAH) is an obstructive pulmonary vasculopathy that results in death from right ventricular failure (RVF). There is limited understanding of the molecular mechanisms of RVF in PAH. Methods: In a PAH-RVF model induced by injection of adult male rats with monocrotaline (MCT; 60 mg/kg), we performed mass spectrometry to identify proteins that change in the RV as a consequence of PAH induced RVF. Bioinformatic analysis was used to integrate our previously published RNA sequencing data from an independent cohort of PAH rats. Results: We identified 1,277 differentially regulated proteins in the RV of MCT rats compared to controls. Integration of MCT RV transcriptome and proteome data sets identified 410 targets that are concordantly regulated at the mRNA and protein levels. Functional analysis of these data revealed enriched functions, including mitochondrial metabolism, cellular respiration, and purine metabolism. We also prioritized 15 highly enriched protein:transcript pairs and confirmed their biological plausibility as contributors to RVF. We demonstrated an overlap of these differentially expressed pairs with data published by independent investigators using multiple PAH models, including the male SU5416-hypoxia model and several male rat strains. Conclusion: Multiomic integration provides a novel view of the molecular phenotype of RVF in PAH which includes dysregulation of pathways involving purine metabolism, mitochondrial function, inflammation, and fibrosis.
Collapse
Affiliation(s)
- Charles Colin Thomas Hindmarch
- QCPU, Queen’s Cardiopulmonary Unit, Translational Institute of Medicine (TIME), Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Lian Tian
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Ping Yu Xiong
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Francois Potus
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et Pneumologie de Quebec, Quebec City, QC, Canada
| | | | - Ruaa Al-Qazazi
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Kurt W. Prins
- Cardiovascular Division, Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Stephen L. Archer
- QCPU, Queen’s Cardiopulmonary Unit, Translational Institute of Medicine (TIME), Department of Medicine, Queen’s University, Kingston, ON, Canada,Department of Medicine, Queen’s University, Kingston, ON, Canada,*Correspondence: Stephen L. Archer,
| |
Collapse
|
9
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
10
|
van der Pijl RJ, Domenighetti AA, Sheikh F, Ehler E, Ottenheijm CAC, Lange S. The titin N2B and N2A regions: biomechanical and metabolic signaling hubs in cross-striated muscles. Biophys Rev 2021; 13:653-677. [PMID: 34745373 PMCID: PMC8553726 DOI: 10.1007/s12551-021-00836-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
Muscle specific signaling has been shown to originate from myofilaments and their associated cellular structures, including the sarcomeres, costameres or the cardiac intercalated disc. Two signaling hubs that play important biomechanical roles for cardiac and/or skeletal muscle physiology are the N2B and N2A regions in the giant protein titin. Prominent proteins associated with these regions in titin are chaperones Hsp90 and αB-crystallin, members of the four-and-a-half LIM (FHL) and muscle ankyrin repeat protein (Ankrd) families, as well as thin filament-associated proteins, such as myopalladin. This review highlights biological roles and properties of the titin N2B and N2A regions in health and disease. Special emphasis is placed on functions of Ankrd and FHL proteins as mechanosensors that modulate muscle-specific signaling and muscle growth. This region of the sarcomere also emerged as a hotspot for the modulation of passive muscle mechanics through altered titin phosphorylation and splicing, as well as tethering mechanisms that link titin to the thin filament system.
Collapse
Affiliation(s)
| | - Andrea A. Domenighetti
- Shirley Ryan AbilityLab, Chicago, IL USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL USA
| | - Farah Sheikh
- Division of Cardiology, School of Medicine, UC San Diego, La Jolla, CA USA
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics, School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| | - Coen A. C. Ottenheijm
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ USA
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Stephan Lange
- Division of Cardiology, School of Medicine, UC San Diego, La Jolla, CA USA
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
11
|
Filomena MC, Yamamoto DL, Carullo P, Medvedev R, Ghisleni A, Piroddi N, Scellini B, Crispino R, D'Autilia F, Zhang J, Felicetta A, Nemska S, Serio S, Tesi C, Catalucci D, Linke WA, Polishchuk R, Poggesi C, Gautel M, Bang ML. Myopalladin knockout mice develop cardiac dilation and show a maladaptive response to mechanical pressure overload. eLife 2021; 10:e58313. [PMID: 34558411 PMCID: PMC8547954 DOI: 10.7554/elife.58313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Myopalladin (MYPN) is a striated muscle-specific immunoglobulin domain-containing protein located in the sarcomeric Z-line and I-band. MYPN gene mutations are causative for dilated (DCM), hypertrophic, and restrictive cardiomyopathy. In a yeast two-hybrid screening, MYPN was found to bind to titin in the Z-line, which was confirmed by microscale thermophoresis. Cardiac analyses of MYPN knockout (MKO) mice showed the development of mild cardiac dilation and systolic dysfunction, associated with decreased myofibrillar isometric tension generation and increased resting tension at longer sarcomere lengths. MKO mice exhibited a normal hypertrophic response to transaortic constriction (TAC), but rapidly developed severe cardiac dilation and systolic dysfunction, associated with fibrosis, increased fetal gene expression, higher intercalated disc fold amplitude, decreased calsequestrin-2 protein levels, and increased desmoplakin and SORBS2 protein levels. Cardiomyocyte analyses showed delayed Ca2+ release and reuptake in unstressed MKO mice as well as reduced Ca2+ spark amplitude post-TAC, suggesting that altered Ca2+ handling may contribute to the development of DCM in MKO mice.
Collapse
Affiliation(s)
- Maria Carmela Filomena
- Institute of Genetic and Biomedical Research (IRGB) - National Research Council (CNR), Milan unitMilanItaly
- IRCCS Humanitas Research HospitalMilanItaly
| | - Daniel L Yamamoto
- Institute of Genetic and Biomedical Research (IRGB) - National Research Council (CNR), Milan unitMilanItaly
| | - Pierluigi Carullo
- Institute of Genetic and Biomedical Research (IRGB) - National Research Council (CNR), Milan unitMilanItaly
- IRCCS Humanitas Research HospitalMilanItaly
| | - Roman Medvedev
- IRCCS Humanitas Research HospitalMilanItaly
- Department of Cardiac Surgery, University of VeronaVeronaItaly
| | - Andrea Ghisleni
- Randall Centre for Cell and Molecular Biophysics, King's College London BHF Centre of Research ExcellenceLondonUnited Kingdom
| | - Nicoletta Piroddi
- Department of Experimental and Clinical Medicine, University of FlorenceFlorenceItaly
| | - Beatrice Scellini
- Department of Experimental and Clinical Medicine, University of FlorenceFlorenceItaly
| | - Roberta Crispino
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
| | | | - Jianlin Zhang
- Department of Medicine, University of California, San DiegoLa JollaUnited States
| | - Arianna Felicetta
- IRCCS Humanitas Research HospitalMilanItaly
- Humanitas UniversityPieve EmanueleItaly
| | | | | | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of FlorenceFlorenceItaly
| | | | - Wolfgang A Linke
- Institute of Physiology II, University of MuensterMuensterGermany
| | - Roman Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of FlorenceFlorenceItaly
| | - Mathias Gautel
- Randall Centre for Cell and Molecular Biophysics, King's College London BHF Centre of Research ExcellenceLondonUnited Kingdom
| | - Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB) - National Research Council (CNR), Milan unitMilanItaly
- IRCCS Humanitas Research HospitalMilanItaly
| |
Collapse
|
12
|
Adewale AO, Ahn YH. Titin N2A Domain and Its Interactions at the Sarcomere. Int J Mol Sci 2021; 22:ijms22147563. [PMID: 34299183 PMCID: PMC8305307 DOI: 10.3390/ijms22147563] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/16/2022] Open
Abstract
Titin is a giant protein in the sarcomere that plays an essential role in muscle contraction with actin and myosin filaments. However, its utility goes beyond mechanical functions, extending to versatile and complex roles in sarcomere organization and maintenance, passive force, mechanosensing, and signaling. Titin’s multiple functions are in part attributed to its large size and modular structures that interact with a myriad of protein partners. Among titin’s domains, the N2A element is one of titin’s unique segments that contributes to titin’s functions in compliance, contraction, structural stability, and signaling via protein–protein interactions with actin filament, chaperones, stress-sensing proteins, and proteases. Considering the significance of N2A, this review highlights structural conformations of N2A, its predisposition for protein–protein interactions, and its multiple interacting protein partners that allow the modulation of titin’s biological effects. Lastly, the nature of N2A for interactions with chaperones and proteases is included, presenting it as an important node that impacts titin’s structural and functional integrity.
Collapse
|
13
|
Affiliation(s)
- Robbert J van der Pijl
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| | - Coen A C Ottenheijm
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| |
Collapse
|
14
|
van der Pijl RJ, van den Berg M, van de Locht M, Shen S, Bogaards SJP, Conijn S, Langlais P, Hooijman PE, Labeit S, Heunks LMA, Granzier H, Ottenheijm CAC. Muscle ankyrin repeat protein 1 (MARP1) locks titin to the sarcomeric thin filament and is a passive force regulator. J Gen Physiol 2021; 153:212403. [PMID: 34152365 PMCID: PMC8222902 DOI: 10.1085/jgp.202112925] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/19/2021] [Indexed: 12/12/2022] Open
Abstract
Muscle ankyrin repeat protein 1 (MARP1) is frequently up-regulated in stressed muscle, but its effect on skeletal muscle function is poorly understood. Here, we focused on its interaction with the titin–N2A element, found in titin’s molecular spring region. We show that MARP1 binds to F-actin, and that this interaction is stronger when MARP1 forms a complex with titin–N2A. Mechanics and super-resolution microscopy revealed that MARP1 “locks” titin–N2A to the sarcomeric thin filament, causing increased extension of titin’s elastic PEVK element and, importantly, increased passive force. In support of this mechanism, removal of thin filaments abolished the effect of MARP1 on passive force. The clinical relevance of this mechanism was established in diaphragm myofibers of mechanically ventilated rats and of critically ill patients. Thus, MARP1 regulates passive force by locking titin to the thin filament. We propose that in stressed muscle, this mechanism protects the sarcomere from mechanical damage.
Collapse
Affiliation(s)
- Robbert J van der Pijl
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tuscon, AZ
| | - Marloes van den Berg
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tuscon, AZ
| | - Martijn van de Locht
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Shengyi Shen
- Department of Cellular and Molecular Medicine, University of Arizona, Tuscon, AZ
| | - Sylvia J P Bogaards
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Stefan Conijn
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Paul Langlais
- Division of Endocrinology, University of Arizona, Tucson, AZ
| | - Pleuni E Hooijman
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Siegfried Labeit
- Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Leo M A Heunks
- Intensive Care Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tuscon, AZ
| | - Coen A C Ottenheijm
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tuscon, AZ
| |
Collapse
|
15
|
Zhou T, Fleming JR, Lange S, Hessel AL, Bogomolovas J, Stronczek C, Grundei D, Ghassemian M, Biju A, Börgeson E, Bullard B, Linke WA, Chen J, Kovermann M, Mayans O. Molecular Characterisation of Titin N2A and Its Binding of CARP Reveals a Titin/Actin Cross-linking Mechanism. J Mol Biol 2021; 433:166901. [PMID: 33647290 PMCID: PMC8052292 DOI: 10.1016/j.jmb.2021.166901] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/14/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022]
Abstract
Striated muscle responds to mechanical overload by rapidly up-regulating the expression of the cardiac ankyrin repeat protein, CARP, which then targets the sarcomere by binding to titin N2A in the I-band region. To date, the role of this interaction in the stress response of muscle remains poorly understood. Here, we characterise the molecular structure of the CARP-receptor site in titin (UN2A) and its binding of CARP. We find that titin UN2A contains a central three-helix bundle fold (ca 45 residues in length) that is joined to N- and C-terminal flanking immunoglobulin domains by long, flexible linkers with partial helical content. CARP binds titin by engaging an α-hairpin in the three-helix fold of UN2A, the C-terminal linker sequence, and the BC loop in Ig81, which jointly form a broad binding interface. Mutagenesis showed that the CARP/N2A association withstands sequence variations in titin N2A and we use this information to evaluate 85 human single nucleotide variants. In addition, actin co-sedimentation, co-transfection in C2C12 cells, proteomics on heart lysates, and the mechanical response of CARP-soaked myofibrils imply that CARP induces the cross-linking of titin and actin myofilaments, thereby increasing myofibril stiffness. We conclude that CARP acts as a regulator of force output in the sarcomere that preserves muscle mechanical performance upon overload stress.
Collapse
Affiliation(s)
- Tiankun Zhou
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | | | - Stephan Lange
- Division of Cardiology, School of Medicine, University of California, San Diego 92093, CA, USA; Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg 413 45, Sweden
| | - Anthony L Hessel
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Julius Bogomolovas
- School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Chiara Stronczek
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - David Grundei
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Majid Ghassemian
- Department of Chemistry and Biochemistry, University of California, San Diego 92093, CA, USA
| | - Andrea Biju
- Division of Cardiology, School of Medicine, University of California, San Diego 92093, CA, USA
| | - Emma Börgeson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg 413 45, Sweden
| | - Belinda Bullard
- Department of Biology, University of York, York YO10 5DD, UK
| | - Wolfgang A Linke
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Ju Chen
- School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael Kovermann
- Department of Chemistry, University of Konstanz, 78457 Konstanz, Germany.
| | - Olga Mayans
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany.
| |
Collapse
|
16
|
Piroddi N, Pesce P, Scellini B, Manzini S, Ganzetti GS, Badi I, Menegollo M, Cora V, Tiso S, Cinquetti R, Monti L, Chiesa G, Bleyl SB, Busnelli M, Dellera F, Bruno D, Caicci F, Grimaldi A, Taramelli R, Manni L, Sacerdoti D, Tesi C, Poggesi C, Ausoni S, Acquati F, Campione M. Myocardial overexpression of ANKRD1 causes sinus venosus defects and progressive diastolic dysfunction. Cardiovasc Res 2021; 116:1458-1472. [PMID: 31688894 DOI: 10.1093/cvr/cvz291] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 09/26/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Abstract
AIMS Increased Ankyrin Repeat Domain 1 (ANKRD1) levels linked to gain of function mutations have been associated to total anomalous pulmonary venous return and adult cardiomyopathy occurrence in humans. The link between increased ANKRD1 level and cardiac structural and functional disease is not understood. To get insight into this problem, we have generated a gain of function ANKRD1 mouse model by overexpressing ANKRD1 in the myocardium. METHODS AND RESULTS Ankrd1 is expressed non-homogeneously in the embryonic myocardium, with a dynamic nucleo-sarcomeric localization in developing cardiomyocytes. ANKRD1 transgenic mice present sinus venosus defect, which originates during development by impaired remodelling of early embryonic heart. Adult transgenic hearts develop diastolic dysfunction with preserved ejection fraction, which progressively evolves into heart failure, as shown histologically and haemodynamically. Transgenic cardiomyocyte structure, sarcomeric assembly, and stability are progressively impaired from embryonic to adult life. Postnatal transgenic myofibrils also present characteristic functional alterations: impaired compliance at neonatal stage and impaired lusitropism in adult hearts. Altogether, our combined analyses suggest that impaired embryonic remodelling and adult heart dysfunction in ANKRD1 transgenic mice present a common ground of initial cardiomyocyte defects, which are exacerbated postnatally. Molecular analysis showed transient activation of GATA4-Nkx2.5 transcription in early transgenic embryos and subsequent dynamic transcriptional modulation within titin gene. CONCLUSIONS ANKRD1 is a fine mediator of cardiomyocyte response to haemodynamic load in the developing and adult heart. Increased ANKRD1 levels are sufficient to initiate an altered cellular phenotype, which is progressively exacerbated into a pathological organ response by the high ventricular workload during postnatal life. Our study defines for the first time a unifying picture for ANKRD1 role in heart development and disease and provides the first mechanistic link between ANKRD1 overexpression and cardiac disease onset.
Collapse
Affiliation(s)
- Nicoletta Piroddi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Paola Pesce
- Department of Medicine, University of Padua, 35121 Padua, Italy
| | - Beatrice Scellini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Giulia S Ganzetti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Ileana Badi
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy.,Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Michela Menegollo
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Virginia Cora
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Simone Tiso
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Raffaella Cinquetti
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Laura Monti
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Steven B Bleyl
- Department of Pediatrics, University of Utah, Salt Lake City, 84132 UT, USA
| | - Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Federica Dellera
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Daniele Bruno
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Federico Caicci
- Department of Biology, University of Padua, 35121 Padua, Italy
| | - Annalisa Grimaldi
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Roberto Taramelli
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Lucia Manni
- Department of Biology, University of Padua, 35121 Padua, Italy
| | - David Sacerdoti
- Department of Medicine, University of Padua, 35121 Padua, Italy
| | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Simonetta Ausoni
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Francesco Acquati
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Marina Campione
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy.,CNR-Neuroscience Institute, 35121 Padua, Italy
| |
Collapse
|
17
|
Yu J, Cai P, Chen X. Structural Regulation of Myocytes in Engineered Healthy and Diseased Cardiac Models. ACS APPLIED BIO MATERIALS 2021; 4:267-276. [DOI: 10.1021/acsabm.0c01270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jing Yu
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Pingqiang Cai
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Xiaodong Chen
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| |
Collapse
|
18
|
Ramachandran CD, Gholami K, Lam SK, Hoe SZ. A preliminary study of the effect of a high-salt diet on transcriptome dynamics in rat hypothalamic forebrain and brainstem cardiovascular control centers. PeerJ 2020; 8:e8528. [PMID: 32175184 PMCID: PMC7059759 DOI: 10.7717/peerj.8528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 01/07/2020] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND High dietary salt intake is strongly correlated with cardiovascular (CV) diseases and it is regarded as a major risk factor associated with the pathogenesis of hypertension. The CV control centres in the brainstem (the nucleus tractus solitarii (NTS) and the rostral ventrolateral medulla (RVLM)) and hypothalamic forebrain (the subfornical organ, SFO; the supraoptic nucleus, SON and the paraventricular nucleus, PVN) have critical roles in regulating CV autonomic motor outflows, and thus maintaining blood pressure (BP). Growing evidence has implicated autonomic regulatory networks in salt-sensitive HPN (SSH), but the genetic basis remains to be delineated. We hypothesized that the development and/ or maintenance of SSH is reliant on the change in the expression of genes in brain regions controlling the CV system. METHODOLOGY We used RNA-Sequencing (RNA-Seq) to describe the differential expression of genes in SFO, SON, PVN, NTS and RVLM of rats being chronically fed with high-salt (HS) diet. Subsequently, a selection of putatively regulated genes was validated with quantitative reverse transcription polymerase chain reaction (qRT-PCR) in both Spontaneously Hypertensive rats (SHRs) and Wistar Kyoto (WKY) rats. RESULTS The findings enabled us to identify number of differentially expressed genes in SFO, SON, PVN, NTS and RVLM; that are either up-regulated in both strains of rats (SON- Caprin2, Sctr), down-regulated in both strains of rats (PVN- Orc, Gkap1), up-regulated only in SHRs (SFO- Apopt1, Lin52, AVP, OXT; SON- AVP, OXT; PVN- Caprin2, Sclt; RVLM- A4galt, Slc29a4, Cmc1) or down-regulated only in SHRs (SON- Ndufaf2, Kcnv1; PVN- Pi4k2a; NTS- Snrpd2l, Ankrd29, St6galnac6, Rnf157, Iglon5, Csrnp3, Rprd1a; RVLM- Ttr, Faim). CONCLUSIONS These findings demonstrated the adverse effects of HS diet on BP, which may be mediated via modulating the signaling systems in CV centers in the hypothalamic forebrain and brainstem.
Collapse
Affiliation(s)
- Chitra Devi Ramachandran
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Perseketuan, Malaysia
| | - Khadijeh Gholami
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Perseketuan, Malaysia
- Human Biology Division, School of Medicine, International Medical University, Kuala Lumpur, Wilayah Perseketuan, Malaysia
| | - Sau Kuen Lam
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Perseketuan, Malaysia
- Department of Pre-Clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long, Selangor, Malaysia
| | - See Ziau Hoe
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Perseketuan, Malaysia
| |
Collapse
|
19
|
Filomena MC, Yamamoto DL, Caremani M, Kadarla VK, Mastrototaro G, Serio S, Vydyanath A, Mutarelli M, Garofalo A, Pertici I, Knöll R, Nigro V, Luther PK, Lieber RL, Beck MR, Linari M, Bang M. Myopalladin promotes muscle growth through modulation of the serum response factor pathway. J Cachexia Sarcopenia Muscle 2020; 11:169-194. [PMID: 31647200 PMCID: PMC7015241 DOI: 10.1002/jcsm.12486] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/01/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Myopalladin (MYPN) is a striated muscle-specific, immunoglobulin-containing protein located in the Z-line and I-band of the sarcomere as well as the nucleus. Heterozygous MYPN gene mutations are associated with hypertrophic, dilated, and restrictive cardiomyopathy, and homozygous loss-of-function truncating mutations have recently been identified in patients with cap myopathy, nemaline myopathy, and congenital myopathy with hanging big toe. METHODS Constitutive MYPN knockout (MKO) mice were generated, and the role of MYPN in skeletal muscle was studied through molecular, cellular, biochemical, structural, biomechanical, and physiological studies in vivo and in vitro. RESULTS MKO mice were 13% smaller compared with wild-type controls and exhibited a 48% reduction in myofibre cross-sectional area (CSA) and significantly increased fibre number. Similarly, reduced myotube width was observed in MKO primary myoblast cultures. Biomechanical studies showed reduced isometric force and power output in MKO mice as a result of the reduced CSA, whereas the force developed by each myosin molecular motor was unaffected. While the performance by treadmill running was similar in MKO and wild-type mice, MKO mice showed progressively decreased exercise capability, Z-line damage, and signs of muscle regeneration following consecutive days of downhill running. Additionally, MKO muscle exhibited progressive Z-line widening starting from 8 months of age. RNA-sequencing analysis revealed down-regulation of serum response factor (SRF)-target genes in muscles from postnatal MKO mice, important for muscle growth and differentiation. The SRF pathway is regulated by actin dynamics as binding of globular actin to the SRF-cofactor myocardin-related transcription factor A (MRTF-A) prevents its translocation to the nucleus where it binds and activates SRF. MYPN was found to bind and bundle filamentous actin as well as interact with MRTF-A. In particular, while MYPN reduced actin polymerization, it strongly inhibited actin depolymerization and consequently increased MRTF-A-mediated activation of SRF signalling in myogenic cells. Reduced myotube width in MKO primary myoblast cultures was rescued by transduction with constitutive active SRF, demonstrating that MYPN promotes skeletal muscle growth through activation of the SRF pathway. CONCLUSIONS Myopalladin plays a critical role in the control of skeletal muscle growth through its effect on actin dynamics and consequently the SRF pathway. In addition, MYPN is important for the maintenance of Z-line integrity during exercise and aging. These results suggest that muscle weakness in patients with biallelic MYPN mutations may be associated with reduced myofibre CSA and SRF signalling and that the disease phenotype may be aggravated by exercise.
Collapse
Affiliation(s)
- Maria Carmela Filomena
- Institute of Genetic and Biomedical Research (IRGB), Milan UnitNational Research CouncilMilanItaly
- Humanitas Clinical and Research CenterRozzanoMilanItaly
| | - Daniel L. Yamamoto
- Institute of Genetic and Biomedical Research (IRGB), Milan UnitNational Research CouncilMilanItaly
| | - Marco Caremani
- Department of BiologyUniversity of FlorenceSesto FiorentinoFlorenceItaly
| | | | | | - Simone Serio
- Humanitas Clinical and Research CenterRozzanoMilanItaly
| | | | | | - Arcamaria Garofalo
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
- Department of Precision MedicineUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Irene Pertici
- Department of BiologyUniversity of FlorenceSesto FiorentinoFlorenceItaly
| | - Ralph Knöll
- Integrated Cardio Metabolic Centre (ICMC), Myocardial GeneticsKarolinska Institutet, University Hospital, Heart and Vascular ThemeSweden
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases (CVRM), Biopharmaceuticals R&DAstraZenecaMölndalSweden
| | - Vincenzo Nigro
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
- Department of Precision MedicineUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | | | - Richard L. Lieber
- Shirley Ryan AbilityLab and Hines V.A. Medical Center ChicagoChicagoILUSA
- Department of Physical Medicine and RehabilitationNorthwestern UniversityChicagoILUSA
- Department of Orthopaedic SurgeryUniversity of California San DiegoLa JollaCAUSA
| | - Moriah R. Beck
- Department of ChemistryWichita State UniversityWichitaKSUSA
| | - Marco Linari
- Department of BiologyUniversity of FlorenceSesto FiorentinoFlorenceItaly
| | - Marie‐Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), Milan UnitNational Research CouncilMilanItaly
- Humanitas Clinical and Research CenterRozzanoMilanItaly
| |
Collapse
|
20
|
Chagula DB, Rechciński T, Rudnicka K, Chmiela M. Ankyrins in human health and disease - an update of recent experimental findings. Arch Med Sci 2020; 16:715-726. [PMID: 32542072 PMCID: PMC7286341 DOI: 10.5114/aoms.2019.89836] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/25/2018] [Indexed: 12/17/2022] Open
Abstract
Ankyrins are adaptor molecules that in eukaryotic cells form complexes with ion channel proteins, cell adhesion and signalling molecules and components of the cytoskeleton. They play a pivotal role as scaffolding proteins, in the structural anchoring to the muscle membrane, in muscle development, neurogenesis and synapse formation. Dysfunction of ankyrins is implicated in numerous diseases such as hereditary spherocytosis, neurodegeneration of Purkinje cells, cardiac arrhythmia, Brugada syndrome, bipolar disorders and schizophrenia, congenital myopathies and congenital heart disease as well as cancers. Detecting either down- or over-expression of ankyrins and ergo their use as biomarkers can provide a new paradigm in the diagnosis of these diseases. This paper provides an outline of knowledge about the structure of ankyrins, and by making use of recent experimental research studies critically discusses their role in several health disorders. Moreover, therapeutic options utilizing engineered ankyrins, designed ankyrin repeat proteins (DARPins), are discussed.
Collapse
Affiliation(s)
- Damian B. Chagula
- Laboratory of Gastroimmunology, Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Tomasz Rechciński
- Department of Cardiology, Bieganski Regional Speciality Hospital, Medical University of Lodz, Lodz, Poland
| | - Karolina Rudnicka
- Laboratory of Gastroimmunology, Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Magdalena Chmiela
- Laboratory of Gastroimmunology, Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
- Corresponding author: Prof. Magdalena Chmiela Laboratory of Gastroimmunology, Department of Immmunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St, 90-237 Lodz, Poland, E-mail:
| |
Collapse
|
21
|
Ankrd2 in Mechanotransduction and Oxidative Stress Response in Skeletal Muscle: New Cues for the Pathogenesis of Muscular Laminopathies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7318796. [PMID: 31428229 PMCID: PMC6681624 DOI: 10.1155/2019/7318796] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/02/2019] [Accepted: 05/19/2019] [Indexed: 12/11/2022]
Abstract
Ankrd2 (ankyrin repeats containing domain 2) or Arpp (ankyrin repeat, PEST sequence, and proline-rich region) is a member of the muscle ankyrin repeat protein family. Ankrd2 is mostly expressed in skeletal muscle, where it plays an intriguing role in the transcriptional response to stress induced by mechanical stimulation as well as by cellular reactive oxygen species. Our studies in myoblasts from Emery-Dreifuss muscular dystrophy 2, a LMNA-linked disease affecting skeletal and cardiac muscles, demonstrated that Ankrd2 is a lamin A-binding protein and that mutated lamins found in Emery-Dreifuss muscular dystrophy change the dynamics of Ankrd2 nuclear import, thus affecting oxidative stress response. In this review, besides describing the latest advances related to Ankrd2 studies, including novel discoveries on Ankrd2 isoform-specific functions, we report the main findings on the relationship of Ankrd2 with A-type lamins and discuss known and potential mechanisms involving defective Ankrd2-lamin A interplay in the pathogenesis of muscular laminopathies.
Collapse
|
22
|
ERK: A Key Player in the Pathophysiology of Cardiac Hypertrophy. Int J Mol Sci 2019; 20:ijms20092164. [PMID: 31052420 PMCID: PMC6539093 DOI: 10.3390/ijms20092164] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/17/2022] Open
Abstract
Cardiac hypertrophy is an adaptive and compensatory mechanism preserving cardiac output during detrimental stimuli. Nevertheless, long-term stimuli incite chronic hypertrophy and may lead to heart failure. In this review, we analyze the recent literature regarding the role of ERK (extracellular signal-regulated kinase) activity in cardiac hypertrophy. ERK signaling produces beneficial effects during the early phase of chronic pressure overload in response to G protein-coupled receptors (GPCRs) and integrin stimulation. These functions comprise (i) adaptive concentric hypertrophy and (ii) cell death prevention. On the other hand, ERK participates in maladaptive hypertrophy during hypertension and chemotherapy-mediated cardiac side effects. Specific ERK-associated scaffold proteins are implicated in either cardioprotective or detrimental hypertrophic functions. Interestingly, ERK phosphorylated at threonine 188 and activated ERK5 (the big MAPK 1) are associated with pathological forms of hypertrophy. Finally, we examine the connection between ERK activation and hypertrophy in (i) transgenic mice overexpressing constitutively activated RTKs (receptor tyrosine kinases), (ii) animal models with mutated sarcomeric proteins characteristic of inherited hypertrophic cardiomyopathies (HCMs), and (iii) mice reproducing syndromic genetic RASopathies. Overall, the scientific literature suggests that during cardiac hypertrophy, ERK could be a “good” player to be stimulated or a “bad” actor to be mitigated, depending on the pathophysiological context.
Collapse
|
23
|
Semantic Multi-Classifier Systems Identify Predictive Processes in Heart Failure Models across Species. Biomolecules 2018; 8:biom8040158. [PMID: 30486323 PMCID: PMC6315933 DOI: 10.3390/biom8040158] [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: 09/20/2018] [Revised: 11/21/2018] [Accepted: 11/21/2018] [Indexed: 11/29/2022] Open
Abstract
Genetic model organisms have the potential of removing blind spots from the underlying gene regulatory networks of human diseases. Allowing analyses under experimental conditions they complement the insights gained from observational data. An inevitable requirement for a successful trans-species transfer is an abstract but precise high-level characterization of experimental findings. In this work, we provide a large-scale analysis of seven weak contractility/heart failure genotypes of the model organism zebrafish which all share a weak contractility phenotype. In supervised classification experiments, we screen for discriminative patterns that distinguish between observable phenotypes (homozygous mutant individuals) as well as wild-type (homozygous wild-types) and carriers (heterozygous individuals). As the method of choice we use semantic multi-classifier systems, a knowledge-based approach which constructs hypotheses from a predefined vocabulary of high-level terms (e.g., Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways or Gene Ontology (GO) terms). Evaluating these models leads to a compact description of the underlying processes and guides the screening for new molecular markers of heart failure. Furthermore, we were able to independently corroborate the identified processes in Wistar rats.
Collapse
|
24
|
Boskovic S, Marín-Juez R, Jasnic J, Reischauer S, El Sammak H, Kojic A, Faulkner G, Radojkovic D, Stainier DYR, Kojic S. Characterization of zebrafish (Danio rerio) muscle ankyrin repeat proteins reveals their conserved response to endurance exercise. PLoS One 2018; 13:e0204312. [PMID: 30252882 PMCID: PMC6155536 DOI: 10.1371/journal.pone.0204312] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/05/2018] [Indexed: 12/30/2022] Open
Abstract
Muscle proteins with ankyrin repeats (MARPs) ANKRD1 and ANKRD2 are titin-associated proteins with a putative role as transcriptional co-regulators in striated muscle, involved in the cellular response to mechanical, oxidative and metabolic stress. Since many aspects of the biology of MARPs, particularly exact mechanisms of their action, in striated muscle are still elusive, research in this field will benefit from novel animal model system. Here we investigated the MARPs found in zebrafish for protein structure, evolutionary conservation, spatiotemporal expression profiles and response to increased muscle activity. Ankrd1 and Ankrd2 show overall moderate conservation at the protein level, more pronounced in the region of ankyrin repeats, motifs indispensable for their function. The two zebrafish genes, ankrd1a and ankrd1b, counterparts of mammalian ANKRD1/Ankrd1, have different expression profiles during first seven days of development. Mild increase of ankrd1a transcript levels was detected at 72 hpf (1.74±0.24 fold increase relative to 24 hpf time point), while ankrd1b expression was markedly upregulated from 24 hpf onward and peaked at 72 hpf (92.18±36.95 fold increase relative to 24 hpf time point). Spatially, they exhibited non-overlapping expression patterns during skeletal muscle development in trunk (ankrd1a) and tail (ankrd1b) somites. Expression of ankrd2 was barely detectable. Zebrafish MARPs, expressed at a relatively low level in adult striated muscle, were found to be responsive to endurance exercise training consisting of two bouts of 3 hours of forced swimming daily, for five consecutive days. Three hours after the last exercise bout, ankrd1a expression increased in cardiac muscle (6.19±5.05 fold change), while ankrd1b and ankrd2 were upregulated in skeletal muscle (1.97±1.05 and 1.84±0.58 fold change, respectively). This study provides the foundation to establish zebrafish as a novel in vivo model for further investigation of MARPs function in striated muscle.
Collapse
Affiliation(s)
- Srdjan Boskovic
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Rubén Marín-Juez
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jovana Jasnic
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Sven Reischauer
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hadil El Sammak
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ana Kojic
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | | | - Dragica Radojkovic
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Snezana Kojic
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
- * E-mail:
| |
Collapse
|
25
|
Bin L, Li X, Richers B, Streib JE, Hu JW, Taylor P, Leung DYM. Ankyrin repeat domain 1 regulates innate immune responses against herpes simplex virus 1: A potential role in eczema herpeticum. J Allergy Clin Immunol 2018; 141:2085-2093.e1. [PMID: 29371118 DOI: 10.1016/j.jaci.2018.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/16/2017] [Accepted: 01/08/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is a common inflammatory skin disease. A subset of patients with AD are susceptible to disseminated herpes simplex virus (HSV) infection, a complication termed eczema herpeticum (ADEH+). The immune mechanisms causing ADEH+ remain elusive. Using RNA sequencing, we recently found that ankyrin repeat domain 1 (ANKRD1) was significantly induced in human PBMCs upon HSV-1 stimulation, and its induction in patients with ADEH+ was significantly reduced compared with that seen in AD patients without a history of eczema herpeticum (ADEH-). OBJECTIVE We sought to validate ANKRD1 gene expression in nonatopic (NA) subjects, patients with ADEH-, and patients with ADEH+ and to delineate the biological function of ANKRD1 and the signaling pathway or pathways involved. METHODS Purification of human PBMCs, monocytes, B cells, dendritic cells, T cells, and natural killer cells; RNA extraction and quantitative RT-PCR; small interfering RNA technique; co-immunoprecipitation; and Western blot assays were used. RESULTS ANKRD1 expression was significantly reduced in PBMCs from patients with ADEH+ after HSV-1 stimulation compared with PBMCs from patients with ADEH-. We found that the induction of ANKRD1 by HSV-1 and multiple pattern recognition receptor agonists are mediated by inflammatory cytokines. Silencing ANKRD1 gene expression in antigen-presenting cells led to increased viral load and reduced IFNB1 and IL29 production. Using co-immunoprecipitation methods, we demonstrated that ANKRD1 formed protein complexes with interferon regulatory factor (IRF) 3 and IRF7, which are important transcription factors regulating signaling transduction of pattern recognition receptors. Overexpression of ANKRD1 enhanced the IRF3-mediated signaling pathways. CONCLUSION ANKRD1 is involved in IRF3-mediated antiviral innate immune signaling pathways. Its reduced expression in patients with ADEH+ might contribute to the pathogenesis of ADEH+.
Collapse
Affiliation(s)
- Lianghua Bin
- Department of Pediatrics, National Jewish Health, Denver, Colo; First Affiliated Hospital, Biomedical Translational Research Institute, the International Immunology Center and the Key Laboratory of Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, China
| | - Xiaozhao Li
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | | | - Joanne E Streib
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | | | - Patricia Taylor
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Donald Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, Colo; University of Colorado School of Medicine, Aurora, Colo.
| |
Collapse
|
26
|
Al Abri MA, Posbergh C, Palermo K, Sutter NB, Eberth J, Hoffman GE, Brooks SA. Genome-Wide Scans Reveal a Quantitative Trait Locus for Withers Height in Horses Near the ANKRD1 Gene. J Equine Vet Sci 2018. [DOI: 10.1016/j.jevs.2017.05.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
27
|
Ankyrin Repeat Domain 1 Protein: A Functionally Pleiotropic Protein with Cardiac Biomarker Potential. Int J Mol Sci 2017; 18:ijms18071362. [PMID: 28672880 PMCID: PMC5535855 DOI: 10.3390/ijms18071362] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 12/20/2022] Open
Abstract
The ankyrin repeat domain 1 (ANKRD1) protein is a cardiac-specific stress-response protein that is part of the muscle ankyrin repeat protein family. ANKRD1 is functionally pleiotropic, playing pivotal roles in transcriptional regulation, sarcomere assembly and mechano-sensing in the heart. Importantly, cardiac ANKRD1 has been shown to be highly induced in various cardiomyopathies and in heart failure, although it is still unclear what impact this may have on the pathophysiology of heart failure. This review aims at highlighting the known properties, functions and regulation of ANKRD1, with focus on the underlying mechanisms that may be involved. The current views on the actions of ANKRD1 in cardiovascular disease and its utility as a candidate cardiac biomarker with diagnostic and/or prognostic potential are also discussed. More studies of ANKRD1 are warranted to obtain deeper functional insights into this molecule to allow assessment of its potential clinical applications as a diagnostic or prognostic marker and/or as a possible therapeutic target.
Collapse
|
28
|
Abstract
Cardiac and skeletal striated muscles are intricately designed machines responsible for muscle contraction. Coordination of the basic contractile unit, the sarcomere, and the complex cytoskeletal networks are critical for contractile activity. The sarcomere is comprised of precisely organized individual filament systems that include thin (actin), thick (myosin), titin, and nebulin. Connecting the sarcomere to other organelles (e.g., mitochondria and nucleus) and serving as the scaffold to maintain cellular integrity are the intermediate filaments. The costamere, on the other hand, tethers the sarcomere to the cell membrane. Unique structures like the intercalated disc in cardiac muscle and the myotendinous junction in skeletal muscle help synchronize and transmit force. Intense investigation has been done on many of the proteins that make up these cytoskeletal assemblies. Yet the details of their function and how they interconnect have just started to be elucidated. A vast number of human myopathies are contributed to mutations in muscle proteins; thus understanding their basic function provides a mechanistic understanding of muscle disorders. In this review, we highlight the components of striated muscle with respect to their interactions, signaling pathways, functions, and connections to disease. © 2017 American Physiological Society. Compr Physiol 7:891-944, 2017.
Collapse
Affiliation(s)
- Christine A Henderson
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Christopher G Gomez
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Stefanie M Novak
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Lei Mi-Mi
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
29
|
Wette SG, Smith HK, Lamb GD, Murphy RM. Characterization of muscle ankyrin repeat proteins in human skeletal muscle. Am J Physiol Cell Physiol 2017; 313:C327-C339. [PMID: 28615162 DOI: 10.1152/ajpcell.00077.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/06/2017] [Accepted: 06/13/2017] [Indexed: 12/23/2022]
Abstract
Muscle ankyrin repeat proteins (MARPs) are a family of titin-associated, stress-response molecules and putative transducers of stretch-induced signaling in skeletal muscle. In cardiac muscle, cardiac ankyrin repeat protein (CARP) and diabetes-related ankyrin repeat protein (DARP) reportedly redistribute from binding sites on titin to the nucleus following a prolonged stretch. However, it is unclear whether ankyrin repeat domain protein 2 (Ankrd 2) shows comparable stretch-induced redistribution to the nucleus. We measured the following in rested human skeletal muscle: 1) the absolute amount of MARPs and 2) the distribution of Ankrd 2 and DARP in both single fibers and whole muscle preparations. In absolute amounts, Ankrd 2 is the most abundant MARP in human skeletal muscle, there being ~3.1 µmol/kg, much greater than DARP and CARP (~0.11 and ~0.02 µmol/kg, respectively). All DARP was found to be tightly bound at cytoskeletal (or possibly nuclear) sites. In contrast, ~70% of the total Ankrd 2 is freely diffusible in the cytosol [including virtually all of the phosphorylated (p)Ankrd 2-Ser99 form], ~15% is bound to non-nuclear membranes, and ~15% is bound at cytoskeletal sites, likely at the N2A region of titin. These data are not consistent with the proposal that Ankrd 2, per se, or pAnkrd 2-Ser99 mediates stretch-induced signaling in skeletal muscle, dissociating from titin and translocating to the nucleus, because the majority of these forms of Ankrd 2 are already free in the cytosol. It will be necessary to show that the titin-associated Ankrd 2 is modified by stretch in some as-yet-unidentified way, distinct from the diffusible pool, if it is to act as a stretch-sensitive signaling molecule.
Collapse
Affiliation(s)
- Stefan G Wette
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Heather K Smith
- Department of Exercise Sciences, The University of Auckland, Auckland, New Zealand
| | - Graham D Lamb
- School of Life Sciences, La Trobe University, Melbourne, Victoria, Australia; and
| | - Robyn M Murphy
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia;
| |
Collapse
|
30
|
Zhang N, Xie XJ, Wang JA. Multifunctional protein: cardiac ankyrin repeat protein. J Zhejiang Univ Sci B 2017; 17:333-41. [PMID: 27143260 DOI: 10.1631/jzus.b1500247] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cardiac ankyrin repeat protein (CARP) not only serves as an important component of muscle sarcomere in the cytoplasm, but also acts as a transcription co-factor in the nucleus. Previous studies have demonstrated that CARP is up-regulated in some cardiovascular disorders and muscle diseases; however, its role in these diseases remains controversial now. In this review, we will discuss the continued progress in the research related to CARP, including its discovery, structure, and the role it plays in cardiac development and heart diseases.
Collapse
Affiliation(s)
- Na Zhang
- Cardiovascular Key Lab of Zhejiang Province, Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiao-Jie Xie
- Cardiovascular Key Lab of Zhejiang Province, Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Jian-An Wang
- Cardiovascular Key Lab of Zhejiang Province, Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
31
|
Zhang N, Ye F, Zhu W, Hu D, Xiao C, Nan J, Su S, Wang Y, Liu M, Gao K, Hu X, Chen J, Yu H, Xie X, Wang J. Cardiac ankyrin repeat protein attenuates cardiomyocyte apoptosis by upregulation of Bcl-2 expression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:3040-3049. [PMID: 27713078 DOI: 10.1016/j.bbamcr.2016.09.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 09/29/2016] [Accepted: 09/30/2016] [Indexed: 12/11/2022]
Abstract
Cardiac ankyrin repeat protein (CARP) is a nuclear transcriptional co-factor that has additional functions in the myoplasm as a component of the muscle sarcomere. Previous studies have demonstrated increased expression of CARP in cardiovascular diseases, however, its role in cardiomyocyte apoptosis is unclear and controversial. In the present study, we investigated possible roles of CARP in hypoxia/reoxygenation (H/R) -induced cardiomyocyte apoptosis and the underlying mechanisms. Neonatal mouse ventricular cardiomyocytes were isolated and infected with adenovirus encoding Flag-tagged CARP (Ad-CARP) and lentivirus encoding CARP targeted shRNA (sh-CARP), respectively. Cardiomyocyte apoptosis induced by exposure to H/R conditions was evaluated by TUNEL staining and western blot analysis of cleaved caspase-3. The results showed that H/R-induced apoptosis was significantly decreased in Ad-CARP cardiomyocytes and increased in sh-CARP cardiomyocytes, suggesting a protective anti-apoptosis role for CARP. Interestingly, over-expressed CARP was mainly distributed in the nucleus, consistent with its role in regulating transcriptional activity. qPCR analysis showed that Bcl-2 transcripts were significantly increased in Ad-CARP cardiomyocytes. ChIP and co-IP assays confirmed the binding of CARP to the Bcl-2 promoter through interaction with transcription factor GATA4. Collectively, our results suggest that CARP can protect against H/R induced cardiomyocyte apoptosis, possibly through increasing anti-apoptosis Bcl-2 gene expression.
Collapse
Affiliation(s)
- Na Zhang
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Feiming Ye
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Wei Zhu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Dexing Hu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Changchen Xiao
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Jinliang Nan
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Sheng'an Su
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Yingchao Wang
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Mingfei Liu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Kanglu Gao
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Xinyang Hu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Jinghai Chen
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, PR China
| | - Hong Yu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Xiaojie Xie
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China.
| | - Jian'an Wang
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China.
| |
Collapse
|
32
|
Zhou T, Fleming JR, Franke B, Bogomolovas J, Barsukov I, Rigden DJ, Labeit S, Mayans O. CARP interacts with titin at a unique helical N2A sequence and at the domain Ig81 to form a structured complex. FEBS Lett 2016; 590:3098-110. [PMID: 27531639 DOI: 10.1002/1873-3468.12362] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/29/2016] [Accepted: 08/09/2016] [Indexed: 11/07/2022]
Abstract
The cardiac ankyrin repeat protein (CARP) is up-regulated in the myocardium during cardiovascular disease and in response to mechanical or toxic stress. Stress-induced CARP interacts with the N2A spring region of the titin filament to modulate muscle compliance. We characterize the interaction between CARP and titin-N2A and show that the binding site in titin spans the dual domain UN2A-Ig81. We find that the unique sequence UN2A is not structurally disordered, but that it has a stable, elongated α-helical fold that possibly acts as a constant force spring. Our findings portray CARP/titin-N2A as a structured node and help to rationalize the molecular basis of CARP mechanosensing in the sarcomeric I-band.
Collapse
Affiliation(s)
- Tiankun Zhou
- Department of Biology, University of Konstanz, Germany.,Institute of Integrative Biology, University of Liverpool, UK
| | - Jennifer R Fleming
- Department of Biology, University of Konstanz, Germany.,Institute of Integrative Biology, University of Liverpool, UK
| | | | - Julius Bogomolovas
- Department of Integrative Pathophysiology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Igor Barsukov
- Institute of Integrative Biology, University of Liverpool, UK
| | - Daniel J Rigden
- Institute of Integrative Biology, University of Liverpool, UK
| | - Siegfried Labeit
- Department of Integrative Pathophysiology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Olga Mayans
- Department of Biology, University of Konstanz, Germany. .,Institute of Integrative Biology, University of Liverpool, UK.
| |
Collapse
|
33
|
Differential expression and localization of Ankrd2 isoforms in human skeletal and cardiac muscles. Histochem Cell Biol 2016; 146:569-584. [PMID: 27393496 DOI: 10.1007/s00418-016-1465-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2016] [Indexed: 01/03/2023]
Abstract
Four human Ankrd2 transcripts, reported in the Ensembl database, code for distinct protein isoforms (360, 333, 327 and 300 aa), and so far, their existence, specific expression and localization patterns have not been studied in detail. Ankrd2 is preferentially expressed in the slow fibers of skeletal muscle. It is found in both the nuclei and the cytoplasm of skeletal muscle cells, and its localization is prone to change during differentiation and upon stress. Ankrd2 has also been detected in the heart, in ventricular cardiomyocytes and in the intercalated disks (ICDs). The main objective of this study was to distinguish between the Ankrd2 isoforms and to determine the contribution of each one to the general profile of Ankrd2 expression in striated muscles. We demonstrated that the known expression and localization pattern of Ankrd2 in striated muscle can be attributed to the isoform of 333 aa which is dominant in both tissues, while the designated cardiac and canonical isoform of 360 aa was less expressed in both tissues. The 360 aa isoform has a distinct nuclear localization in human skeletal muscle, as well as in primary myoblasts and myotubes. In contrast to the isoform of 333 aa, it was not preferentially expressed in slow fibers and not localized to the ICDs of human cardiomyocytes. Regulation of the expression of both isoforms is achieved at the transcriptional level. Our results set the stage for investigation of the specific functions and interactions of the Ankrd2 isoforms in healthy and diseased human striated muscles.
Collapse
|
34
|
Lange S, Gehmlich K, Lun AS, Blondelle J, Hooper C, Dalton ND, Alvarez EA, Zhang X, Bang ML, Abassi YA, Dos Remedios CG, Peterson KL, Chen J, Ehler E. MLP and CARP are linked to chronic PKCα signalling in dilated cardiomyopathy. Nat Commun 2016; 7:12120. [PMID: 27353086 PMCID: PMC4931343 DOI: 10.1038/ncomms12120] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 05/31/2016] [Indexed: 11/28/2022] Open
Abstract
MLP (muscle LIM protein)-deficient mice count among the first mouse models for dilated cardiomyopathy (DCM), yet the exact role of MLP in cardiac signalling processes is still enigmatic. Elevated PKCα signalling activity is known to be an important contributor to heart failure. Here we show that MLP directly inhibits the activity of PKCα. In end-stage DCM, PKCα is concentrated at the intercalated disc of cardiomyocytes, where it is sequestered by the adaptor protein CARP in a multiprotein complex together with PLCβ1. In mice deficient for both MLP and CARP the chronic PKCα signalling chain at the intercalated disc is broken and they remain healthy. Our results suggest that the main role of MLP in heart lies in the direct inhibition of PKCα and that chronic uninhibited PKCα activity at the intercalated disc in the absence of functional MLP leads to heart failure. Altered function of the muscle LIM protein (MLP) causes dilated cardiomyopathy in mice and humans. Lange et al. explain the molecular role of MLP in the heart by showing that it affects the signalling complex at the intercalated discs of failing hearts that consists of PKCα, PLCβ1 and CARP by inhibiting PKCα auto-phosphorylation and function.
Collapse
Affiliation(s)
- Stephan Lange
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Katja Gehmlich
- BHF Centre of Research Excellence Oxford, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Alexander S Lun
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Jordan Blondelle
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Charlotte Hooper
- BHF Centre of Research Excellence Oxford, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Nancy D Dalton
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Erika A Alvarez
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Xiaoyu Zhang
- ACEA Biosciences, 6779 Mesa Ridge Rd #100, San Diego, CA-92121, USA
| | - Marie-Louise Bang
- Institute of Genetic and Biomedical Research, UOS Milan, National Research Council, Rozzano (Milan) 20089, Italy.,Humanitas Clinical and Research Center, Rozzano (Milan) 20089, Italy
| | - Yama A Abassi
- ACEA Biosciences, 6779 Mesa Ridge Rd #100, San Diego, CA-92121, USA
| | | | - Kirk L Peterson
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Ju Chen
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Elisabeth Ehler
- BHF Centre of Research Excellence at King's College London, Cardiovascular Division and Randall Division of Cell and Molecular Biophysics, London SE1 1UL, UK
| |
Collapse
|
35
|
Krüger M, Kötter S. Titin, a Central Mediator for Hypertrophic Signaling, Exercise-Induced Mechanosignaling and Skeletal Muscle Remodeling. Front Physiol 2016; 7:76. [PMID: 26973541 PMCID: PMC4771757 DOI: 10.3389/fphys.2016.00076] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/15/2016] [Indexed: 01/09/2023] Open
Abstract
Titin is a giant scaffold protein with multiple functions in striated muscle physiology. Due to the elastic I-band domains and the filament-like integration in the half-sarcomere titin is an important factor for sarcomere assembly and serves as an adaptable molecular spring that determines myofilament distensibility. Protein-interactions e.g., with muscle ankyrin repeat proteins or muscle LIM-protein link titin to hypertrophic signaling and via p62 and Muscle Ring Finger proteins to mechanisms that control protein quality control. This review summarizes our current knowledge on titin as a central node for exercise-induced mechanosignaling and remodeling and further highlights the pathophysiological implications.
Collapse
Affiliation(s)
- Martina Krüger
- Institute of Cardiovascular Physiology, Heinrich Heine University Düsseldorf Düsseldorf, Germany
| | - Sebastian Kötter
- Institute of Cardiovascular Physiology, Heinrich Heine University Düsseldorf Düsseldorf, Germany
| |
Collapse
|
36
|
Liang Y, Sheikh F. Scaffold Proteins Regulating Extracellular Regulated Kinase Function in Cardiac Hypertrophy and Disease. Front Pharmacol 2016; 7:37. [PMID: 26973524 PMCID: PMC4770026 DOI: 10.3389/fphar.2016.00037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 02/11/2016] [Indexed: 01/07/2023] Open
Abstract
The mitogen activated protein kinase (MAPK)-extracellular regulated kinase 1/2 (ERK1/2) pathway is a central downstream signaling pathway that is activated in cardiac muscle cells during mechanical and agonist-mediated hypertrophy. Studies in genetic mouse models deficient in ERK-associated MAPK components pathway have further reinforced a direct role for this pathway in stress-induced cardiac hypertrophy and disease. However, more recent studies have highlighted that these signaling pathways may exert their regulatory functions in a more compartmentalized manner in cardiac muscle. Emerging data has uncovered specific MAPK scaffolding proteins that tether MAPK/ERK signaling specifically at the sarcomere and plasma membrane in cardiac muscle and show that deficiencies in these scaffolding proteins alter ERK activity and phosphorylation, which are then critical in altering the cardiac myocyte response to stress-induced hypertrophy and disease progression. In this review, we provide insights on ERK-associated scaffolding proteins regulating cardiac myofilament function and their impact on cardiac hypertrophy and disease.
Collapse
Affiliation(s)
- Yan Liang
- Department of Medicine, University of California-San Diego, La Jolla CA, USA
| | - Farah Sheikh
- Department of Medicine, University of California-San Diego, La Jolla CA, USA
| |
Collapse
|
37
|
Ma C, Ying Y, Zhang T, Zhang W, Peng H, Cheng X, Xu L, Tong H. Establishment of a prediction model of changing trends in cardiac hypertrophy disease based on microarray data screening. Exp Ther Med 2016; 11:1734-1740. [PMID: 27168795 PMCID: PMC4840528 DOI: 10.3892/etm.2016.3105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 01/15/2016] [Indexed: 12/13/2022] Open
Abstract
The aim of the present study was to construct a mathematical model to predict the changing trends of cardiac hypertrophy at gene level. Microarray data were downloaded from Gene Expression Omnibus database (accession, GSE21600), which included 35 samples harvested from the heart of Wistar rats on postoperative days 1 (D1 group), 6 (D6 group) and 42 (D42 group) following aorta ligation and sham operated Wistar rats, respectively. Each group contained six samples, with the exception of the samples harvested from the aorta ligated group after 6 days, where n=5. Differentially expressed genes (DEGs) were identified using a Limma package in R. Hierarchical clustering analysis was performed on common DEGs in order to construct a linear equation between the D1 and D42 groups, using linear discriminant analysis. Subsequent verification was performed using receiver operating characteristic (ROC) curve and the measurement data at day 42. A total of 319, 44 and 57 DEGs were detected in D1, D6 and D42 sample groups, respectively. AKIP1, ANKRD23, LTBP2, TGF-β2 and TNFRSF12A were identified as common DEGs in all groups. The predicted linear equation between D1 and D42 group was calculated to be y=1.526×-186.671. Assessment of the ROC curve demonstrated that the area under the curve was 0.831, with a specificity and sensitivity of 0.8. As compared with the predictive and measurement data at day 42, the consistency of the two sets of data was 76.5%. In conclusion, the present model may contribute to the early prediction of changing trends in cardiac hypertrophy disease at gene level.
Collapse
Affiliation(s)
- Caiyan Ma
- Cardiovascular Department, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Yongjun Ying
- Cardiovascular Department, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Tianjie Zhang
- Cardiovascular Department, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Wei Zhang
- Cardiovascular Department, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Hui Peng
- Cardiovascular Department, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Xufeng Cheng
- Cardiovascular Department, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Lin Xu
- Cardiovascular Department, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Hong Tong
- Cardiovascular Department, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| |
Collapse
|
38
|
Shimoda Y, Matsuo K, Kitamura Y, Ono K, Ueyama T, Matoba S, Yamada H, Wu T, Chen J, Emoto N, Ikeda K. Diabetes-Related Ankyrin Repeat Protein (DARP/Ankrd23) Modifies Glucose Homeostasis by Modulating AMPK Activity in Skeletal Muscle. PLoS One 2015; 10:e0138624. [PMID: 26398569 PMCID: PMC4580461 DOI: 10.1371/journal.pone.0138624] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 08/31/2015] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle is the major site for glucose disposal, the impairment of which closely associates with the glucose intolerance in diabetic patients. Diabetes-related ankyrin repeat protein (DARP/Ankrd23) is a member of muscle ankyrin repeat proteins, whose expression is enhanced in the skeletal muscle under diabetic conditions; however, its role in energy metabolism remains poorly understood. Here we report a novel role of DARP in the regulation of glucose homeostasis through modulating AMP-activated protein kinase (AMPK) activity. DARP is highly preferentially expressed in skeletal muscle, and its expression was substantially upregulated during myotube differentiation of C2C12 myoblasts. Interestingly, DARP-/- mice demonstrated better glucose tolerance despite similar body weight, while their insulin sensitivity did not differ from that in wildtype mice. We found that phosphorylation of AMPK, which mediates insulin-independent glucose uptake, in skeletal muscle was significantly enhanced in DARP-/- mice compared to that in wildtype mice. Gene silencing of DARP in C2C12 myotubes enhanced AMPK phosphorylation, whereas overexpression of DARP in C2C12 myoblasts reduced it. Moreover, DARP-silencing increased glucose uptake and oxidation in myotubes, which was abrogated by the treatment with AICAR, an AMPK activator. Of note, improved glucose tolerance in DARP-/- mice was abolished when mice were treated with AICAR. Mechanistically, gene silencing of DARP enhanced protein expression of LKB1 that is a major upstream kinase for AMPK in myotubes in vitro and the skeletal muscle in vivo. Together with the altered expression under diabetic conditions, our data strongly suggest that DARP plays an important role in the regulation of glucose homeostasis under physiological and pathological conditions, and thus DARP is a new therapeutic target for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Yoshiaki Shimoda
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Kiyonari Matsuo
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Youhei Kitamura
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Kazunori Ono
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Tomomi Ueyama
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Satoaki Matoba
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Hiroyuki Yamada
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Tongbin Wu
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Noriaki Emoto
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kitamachi, Higashinada, Kobe6588558, Japan
| | - Koji Ikeda
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kitamachi, Higashinada, Kobe6588558, Japan
- * E-mail:
| |
Collapse
|
39
|
Abstract
The members of the nebulin protein family, including nebulin, nebulette, LASP-1, LASP-2, and N-RAP, contain various numbers of nebulin repeats and bind to actin, but are otherwise heterogeneous with regard to size, expression pattern, and function. This review focuses on the roles of nebulin family members in the heart. Nebulin is the largest member predominantly expressed in skeletal muscle, where it stretches along the thin filament. In heart, nebulin is detectable only at low levels and its absence has no apparent effects. Nebulette is similar in structure to the nebulin C-terminal Z-line region and specifically expressed in heart. Nebulette gene mutations have been identified in dilated cardiomyopathy patients and transgenic mice overexpressing nebulette mutants partially recapitulate the human pathology. In contrast, nebulette knockout mice show no functional phenotype, but exhibit Z-line widening. LASP-2 is an isoform of nebulette expressed in multiple tissues, including the heart. It is present in the Z-line and intercalated disc and able to bind and cross-link filamentous actin. LASP-1 is similar in structure to LASP-2, but expressed only in non-muscle tissue. N-RAP is present in myofibril precursors during myofibrillogenesis and thought to be involved in myofibril assembly, while it is localized at the intercalated disc in adult heart. Additional in vivo models are required to provide further insights into the functions of nebulin family members in the heart.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research, UOS Milan, National Research Council
| | | |
Collapse
|
40
|
Mastrototaro G, Liang X, Li X, Carullo P, Piroddi N, Tesi C, Gu Y, Dalton ND, Peterson KL, Poggesi C, Sheikh F, Chen J, Bang ML. Nebulette knockout mice have normal cardiac function, but show Z-line widening and up-regulation of cardiac stress markers. Cardiovasc Res 2015; 107:216-25. [PMID: 25987543 DOI: 10.1093/cvr/cvv156] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 04/26/2015] [Indexed: 02/06/2023] Open
Abstract
AIMS Nebulette is a 109 kDa modular protein localized in the sarcomeric Z-line of the heart. In vitro studies have suggested a role of nebulette in stabilizing the thin filament, and missense mutations in the nebulette gene were recently shown to be causative for dilated cardiomyopathy and endocardial fibroelastosis in human and mice. However, the role of nebulette in vivo has remained elusive. To provide insights into the function of nebulette in vivo, we generated and studied nebulette-deficient (nebl(-) (/-)) mice. METHODS AND RESULTS Nebl(-) (/-) mice were generated by replacement of exon 1 by Cre under the control of the endogenous nebulette promoter, allowing for lineage analysis using the ROSA26 Cre reporter strain. This revealed specific expression of nebulette in the heart, consistent with in situ hybridization results. Nebl(-) (/-) mice exhibited normal cardiac function both under basal conditions and in response to transaortic constriction as assessed by echocardiography and haemodynamic analyses. Furthermore, histological, IF, and western blot analysis showed no cardiac abnormalities in nebl(-) (/-) mice up to 8 months of age. In contrast, transmission electron microscopy showed Z-line widening starting from 5 months of age, suggesting that nebulette is important for the integrity of the Z-line. Furthermore, up-regulation of cardiac stress responsive genes suggests the presence of chronic cardiac stress in nebl(-) (/-) mice. CONCLUSION Nebulette is dispensable for normal cardiac function, although Z-line widening and up-regulation of cardiac stress markers were found in nebl(-) (/-) heart. These results suggest that the nebulette disease causing mutations have dominant gain-of-function effects.
Collapse
Affiliation(s)
- Giuseppina Mastrototaro
- Humanitas Clinical and Research Center, Via Manzoni 113, 20089 Rozzano, Milan, Italy University of Milano-Bicocca, Milan, Italy
| | - Xingqun Liang
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
| | - Xiaodong Li
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
| | - Pierluigi Carullo
- Humanitas Clinical and Research Center, Via Manzoni 113, 20089 Rozzano, Milan, Italy Institute of Genetic and Biomedical Research, UOS Milan, National Research Council, Milan, Italy
| | - Nicoletta Piroddi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Yusu Gu
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
| | - Nancy D Dalton
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
| | - Kirk L Peterson
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Farah Sheikh
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
| | - Ju Chen
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
| | - Marie-Louise Bang
- Humanitas Clinical and Research Center, Via Manzoni 113, 20089 Rozzano, Milan, Italy Institute of Genetic and Biomedical Research, UOS Milan, National Research Council, Milan, Italy
| |
Collapse
|
41
|
Lyon RC, Zanella F, Omens JH, Sheikh F. Mechanotransduction in cardiac hypertrophy and failure. Circ Res 2015; 116:1462-1476. [PMID: 25858069 PMCID: PMC4394185 DOI: 10.1161/circresaha.116.304937] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/13/2015] [Indexed: 01/10/2023]
Abstract
Cardiac muscle cells have an intrinsic ability to sense and respond to mechanical load through a process known as mechanotransduction. In the heart, this process involves the conversion of mechanical stimuli into biochemical events that induce changes in myocardial structure and function. Mechanotransduction and its downstream effects function initially as adaptive responses that serve as compensatory mechanisms during adaptation to the initial load. However, under prolonged and abnormal loading conditions, the remodeling processes can become maladaptive, leading to altered physiological function and the development of pathological cardiac hypertrophy and heart failure. Although the mechanisms underlying mechanotransduction are far from being fully elucidated, human and mouse genetic studies have highlighted various cytoskeletal and sarcolemmal structures in cardiac myocytes as the likely candidates for load transducers, based on their link to signaling molecules and architectural components important in disease pathogenesis. In this review, we summarize recent developments that have uncovered specific protein complexes linked to mechanotransduction and mechanotransmission within the sarcomere, the intercalated disc, and at the sarcolemma. The protein structures acting as mechanotransducers are the first step in the process that drives physiological and pathological cardiac hypertrophy and remodeling, as well as the transition to heart failure, and may provide better insights into mechanisms driving mechanotransduction-based diseases.
Collapse
Affiliation(s)
- Robert C. Lyon
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Fabian Zanella
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Jeffrey H. Omens
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Farah Sheikh
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| |
Collapse
|
42
|
Zhong L, Chiusa M, Cadar AG, Lin A, Samaras S, Davidson JM, Lim CC. Targeted inhibition of ANKRD1 disrupts sarcomeric ERK-GATA4 signal transduction and abrogates phenylephrine-induced cardiomyocyte hypertrophy. Cardiovasc Res 2015; 106:261-71. [PMID: 25770146 DOI: 10.1093/cvr/cvv108] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 03/05/2015] [Indexed: 12/11/2022] Open
Abstract
AIMS Accumulating evidence suggest that sarcomere signalling complexes play a pivotal role in cardiomyocyte hypertrophy by communicating stress signals to the nucleus to induce gene expression. Ankyrin repeat domain 1 (ANKRD1) is a transcriptional regulatory protein that also associates with sarcomeric titin; however, the exact role of ANKRD1 in the heart remains to be elucidated. We therefore aimed to examine the role of ANKRD1 in cardiomyocyte hypertrophic signalling. METHODS AND RESULTS In neonatal rat ventricular myocytes, we found that ANKRD1 is part of a sarcomeric signalling complex that includes ERK1/2 and cardiac transcription factor GATA4. Treatment with hypertrophic agonist phenylephrine (PE) resulted in phosphorylation of ERK1/2 and GATA4 followed by nuclear translocation of the ANKRD1/ERK/GATA4 complex. Knockdown of Ankrd1 attenuated PE-induced phosphorylation of ERK1/2 and GATA4, inhibited nuclear translocation of the ANKRD1 complex, and prevented cardiomyocyte growth. Mice lacking Ankrd1 are viable with normal cardiac function. Chronic PE infusion in wild-type mice induced significant cardiac hypertrophy with reactivation of the cardiac fetal gene program which was completely abrogated in Ankrd1 null mice. In contrast, ANKRD1 does not play a role in haemodynamic overload as Ankrd1 null mice subjected to transverse aortic constriction developed cardiac hypertrophy comparable to wild-type mice. CONCLUSION Our study reveals a novel role for ANKRD1 as a selective regulator of PE-induced signalling whereby ANKRD1 recruits and localizes GATA4 and ERK1/2 in a sarcomeric macro-molecular complex to enhance GATA4 phosphorylation with subsequent nuclear translocation of the ANKRD1 complex to induce hypertrophic gene expression.
Collapse
Affiliation(s)
- Lin Zhong
- Department of Medicine, Cardiovascular Division, Vanderbilt University School of Medicine, 2220 Pierce Ave, Preston Research Building, Rm 332, Nashville, TN 37232, USA
| | - Manuel Chiusa
- Department of Medicine, Cardiovascular Division, Vanderbilt University School of Medicine, 2220 Pierce Ave, Preston Research Building, Rm 332, Nashville, TN 37232, USA
| | - Adrian G Cadar
- Department of Medicine, Cardiovascular Division, Vanderbilt University School of Medicine, 2220 Pierce Ave, Preston Research Building, Rm 332, Nashville, TN 37232, USA Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Angel Lin
- Department of Medicine, Cardiovascular Division, Vanderbilt University School of Medicine, 2220 Pierce Ave, Preston Research Building, Rm 332, Nashville, TN 37232, USA
| | - Susan Samaras
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jeffrey M Davidson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA Research Service, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37232, USA
| | - Chee C Lim
- Department of Medicine, Cardiovascular Division, Vanderbilt University School of Medicine, 2220 Pierce Ave, Preston Research Building, Rm 332, Nashville, TN 37232, USA Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
43
|
Correction: The muscle ankyrin repeat proteins CARP, Ankrd2, and DARP are not essential for normal cardiac development and function at basal conditions and in response to pressure overload. PLoS One 2015; 10:e0118523. [PMID: 25747160 PMCID: PMC4351946 DOI: 10.1371/journal.pone.0118523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
44
|
Huby AC, Mendsaikhan U, Takagi K, Martherus R, Wansapura J, Gong N, Osinska H, James JF, Kramer K, Saito K, Robbins J, Khuchua Z, Towbin JA, Purevjav E. Disturbance in Z-disk mechanosensitive proteins induced by a persistent mutant myopalladin causes familial restrictive cardiomyopathy. J Am Coll Cardiol 2015; 64:2765-76. [PMID: 25541130 DOI: 10.1016/j.jacc.2014.09.071] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 08/18/2014] [Accepted: 09/04/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Familial restrictive cardiomyopathy (FRCM) has a poor prognosis due to diastolic dysfunction and restrictive physiology (RP). Myocardial stiffness, with or without fibrosis, underlie RP, but the mechanism(s) of restrictive remodeling is unclear. Myopalladin (MYPN) is a messenger molecule that links structural and gene regulatory molecules via translocation from the Z-disk and I-bands to the nucleus in cardiomyocytes. Expression of N-terminal MYPN peptide results in severe disruption of the sarcomere. OBJECTIVES The aim was to study a nonsense MYPN-Q529X mutation previously identified in the FRCM family in an animal model to explore the molecular and pathogenic mechanisms of FRCM. METHODS Functional (echocardiography, cardiac magnetic resonance [CMR] imaging, electrocardiography), morphohistological, gene expression, and molecular studies were performed in knock-in heterozygote (Mypn(WT/Q526X)) and homozygote mice harboring the human MYPN-Q529X mutation. RESULTS Echocardiographic and CMR imaging signs of diastolic dysfunction with preserved systolic function were identified in 12-week-old Mypn(WT/Q526X) mice. Histology revealed interstitial and perivascular fibrosis without overt hypertrophic remodeling. Truncated Mypn(Q526X) protein was found to translocate to the nucleus. Levels of total and nuclear cardiac ankyrin repeat protein (Carp/Ankrd1) and phosphorylation of mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 (Erk1/2), Erk1/2, Smad2, and Akt were reduced. Up-regulation was evident for muscle LIM protein (Mlp), desmin, and heart failure (natriuretic peptide A [Nppa], Nppb, and myosin heavy chain 6) and fibrosis (transforming growth factor beta 1, alpha-smooth muscle actin, osteopontin, and periostin) markers. CONCLUSIONS Heterozygote Mypn(WT/Q526X) knock-in mice develop RCM due to persistence of mutant Mypn(Q526X) protein in the nucleus. Down-regulation of Carp and up-regulation of Mlp and desmin appear to augment fibrotic restrictive remodeling, and reduced Erk1/2 levels blunt a hypertrophic response in Mypn(WT/Q526X) hearts.
Collapse
Affiliation(s)
- Anne-Cecile Huby
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Uzmee Mendsaikhan
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - Ruben Martherus
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Janaka Wansapura
- Department of Radiology, Imaging Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Nan Gong
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Hanna Osinska
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jeanne F James
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kristen Kramer
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kazuyoshi Saito
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jeffrey Robbins
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Zaza Khuchua
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jeffrey A Towbin
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Enkhsaikhan Purevjav
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| |
Collapse
|
45
|
Jasnic-Savovic J, Nestorovic A, Savic S, Karasek S, Vitulo N, Valle G, Faulkner G, Radojkovic D, Kojic S. Profiling of skeletal muscle Ankrd2 protein in human cardiac tissue and neonatal rat cardiomyocytes. Histochem Cell Biol 2015; 143:583-97. [PMID: 25585647 DOI: 10.1007/s00418-015-1307-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2014] [Indexed: 10/24/2022]
Abstract
Muscle-specific mechanosensors Ankrd2/Arpp (ankyrin repeat protein 2) and Ankrd1/CARP (cardiac ankyrin repeat protein) have an important role in transcriptional regulation, myofibrillar assembly, cardiogenesis and myogenesis. In skeletal muscle myofibrils, Ankrd2 has a structural role as a component of a titin associated stretch-sensing complex, while in the nucleus it exerts regulatory function as transcriptional co-factor. It is also involved in myogenic differentiation and coordination of myoblast proliferation. Although expressed in the heart, the role of Ankrd2 in the cardiac muscle is completely unknown. Recently, we have shown that hypertrophic and dilated cardiomyopathy pathways are altered upon Ankrd2 silencing suggesting the importance of this protein in cardiac tissue. Here we provide the underlying basis for the functional investigation of Ankrd2 in the heart. We confirmed reduced Ankrd2 expression levels in human heart in comparison with Ankrd1 using RNAseq and Western blot. For the first time we demonstrated that, apart from the sarcomere and nucleus, both proteins are localized to the intercalated disks of human cardiomyocytes. We further tested the expression and localization of endogenous Ankrd2 in rat neonatal cardiomyocytes, a well-established model for studying cardiac-specific proteins. Ankrd2 was found to be expressed in both the cytoplasm and nucleus, independently from maturation status of cardiomyocytes. In contrast to Ankrd1, it is not responsive to the cardiotoxic drug Doxorubicin, suggesting that different mechanisms govern their expression in cardiac cells.
Collapse
Affiliation(s)
- Jovana Jasnic-Savovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, PO Box 23, 11010, Belgrade, Serbia
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Samaras SE, Almodóvar-García K, Wu N, Yu F, Davidson JM. Global deletion of Ankrd1 results in a wound-healing phenotype associated with dermal fibroblast dysfunction. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:96-109. [PMID: 25452119 DOI: 10.1016/j.ajpath.2014.09.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/14/2014] [Accepted: 09/19/2014] [Indexed: 12/27/2022]
Abstract
The expression of ankyrin repeat domain protein 1 (Ankrd1), a transcriptional cofactor and sarcomeric component, is strongly elevated by wounding and tissue injury. We developed a conditional Ankrd1(fl/fl) mouse, performed global deletion with Sox2-cre, and assessed the role of this protein in cutaneous wound healing. Although global deletion of Ankrd1 did not affect mouse viability or development, Ankrd1(-/-) mice had at least two significant wound-healing phenotypes: extensive necrosis of ischemic skin flaps, which was reversed by adenoviral expression of ANKRD1, and delayed excisional wound closure, which was characterized by decreased contraction and reduced granulation tissue thickness. Skin fibroblasts isolated from Ankrd1(-/-) mice did not spread or migrate on collagen- or fibronectin-coated surfaces as efficiently as fibroblasts isolated from Ankrd1(fl/fl) mice. More important, Ankrd1(-/-) fibroblasts failed to contract three-dimensional floating collagen gels. Reconstitution of ANKRD1 by adenoviral infection stimulated both collagen gel contraction and actin fiber organization. These in vitro data were consistent with in vivo wound closure studies, and suggest that ANKRD1 is important for the proper interaction of fibroblasts with a compliant collagenous matrix both in vitro and in vivo.
Collapse
Affiliation(s)
- Susan E Samaras
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Karinna Almodóvar-García
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Nanjun Wu
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Fang Yu
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Jeffrey M Davidson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee; Research Service, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee.
| |
Collapse
|