1
|
Fujita K, Desmond P, Blondelle J, Soták M, Rajan MR, Clark M, Estève É, Chan Y, Gu Y, Actis Dato V, Marrocco V, Dalton ND, Ghassemian M, Do A, Klos M, Peterson KL, Sheikh F, Cho Y, Börgeson E, Lange S. Combined Loss of Obsc and Obsl1 in Murine Hearts Results in Diastolic Dysfunction, Altered Metabolism, and Deregulated Mitophagy. Circ Heart Fail 2025; 18:e011867. [PMID: 40066567 PMCID: PMC11995854 DOI: 10.1161/circheartfailure.124.011867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 01/09/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Muscle proteins of the obscurin protein family play important roles in sarcomere organization and sarcoplasmic reticulum and T-tubule architecture and function. However, their precise molecular functions and redundancies between protein family members as well as their involvement in cardiac diseases remain to be fully understood. METHODS To investigate the functional roles of Obsc (obscurin) and its close homolog Obsl1 (obscurin-like 1) in the heart, we generated and analyzed knockout mice for Obsc, Obsl1, as well as Obsc/Obsl1 double knockouts. RESULTS We show that double-knockout mice are viable but show postnatal deficits in cardiac muscle sarcoplasmic reticulum and mitochondrial architecture and function at the microscopic, biochemical, and cellular levels. Altered sarcoplasmic reticulum structure resulted in perturbed calcium cycling, whereas mitochondrial ultrastructure deficits were linked to decreased levels of Chchd3 (coiled-coil-helix-coiled-coil-helix domain containing 3), a Micos (mitochondrial contact site and cristae organizing system) complex protein. Hearts of double-knockout mice also show altered levels of Atg4 proteins, novel Obsl1 interactors, resulting in abnormal mitophagy, and increased unfolded protein response. At the physiological level, loss of obscurin and Obsl1 resulted in a profound delay of cardiac relaxation, associated with metabolic signs of heart failure. CONCLUSIONS Taken together, our data suggest that Obsc and Obsl1 play crucial roles in cardiac sarcoplasmic reticulum structure, calcium cycling, mitochondrial function, turnover, and metabolism.
Collapse
Affiliation(s)
- Kyohei Fujita
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Japan (K.F.)
| | - Patrick Desmond
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Jordan Blondelle
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Matúš Soták
- Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, University of Gothenburg, Sweden (M.S., M.R.R., E.B.)
| | - Meenu Rohini Rajan
- Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, University of Gothenburg, Sweden (M.S., M.R.R., E.B.)
| | - Madison Clark
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
- Department of Biomedicine, Aarhus University, Denmark (M.C., E.B., S.L.)
- STENO Diabetes Center Aarhus, Denmark (M.C., E.B., S.L.)
| | - Éric Estève
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
- PhyMedExp, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Regionale Universitaire (CHRU) Montpellier, France (E.E.)
| | - Yunghang Chan
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Yusu Gu
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Virginia Actis Dato
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Valeria Marrocco
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Nancy D. Dalton
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Majid Ghassemian
- Department of Chemistry and Biochemistry (M.G.), University of California San Diego, La Jolla
| | - Aryanne Do
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Matthew Klos
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Kirk L. Peterson
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Farah Sheikh
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Yoshitake Cho
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Emma Börgeson
- Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, University of Gothenburg, Sweden (M.S., M.R.R., E.B.)
- Department of Biomedicine, Aarhus University, Denmark (M.C., E.B., S.L.)
- STENO Diabetes Center Aarhus, Denmark (M.C., E.B., S.L.)
| | - Stephan Lange
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
- Department of Biomedicine, Aarhus University, Denmark (M.C., E.B., S.L.)
- STENO Diabetes Center Aarhus, Denmark (M.C., E.B., S.L.)
| |
Collapse
|
2
|
Granzier HL, Labeit S. Discovery of Titin and Its Role in Heart Function and Disease. Circ Res 2025; 136:135-157. [PMID: 39745989 DOI: 10.1161/circresaha.124.323051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 01/04/2025]
Abstract
This review examines the giant elastic protein titin and its critical roles in heart function, both in health and disease, as discovered since its identification nearly 50 years ago. Encoded by the TTN (titin gene), titin has emerged as a major disease locus for cardiac disorders. Functionally, titin acts as a third myofilament type, connecting sarcomeric Z-disks and M-bands, and regulating myocardial passive stiffness and stretch sensing. Its I-band segment, which includes the N2B element and the PEVK (proline, glutamate, valine, and lysine-rich regions), serves as a viscoelastic spring, adjusting sarcomere length and force in response to cardiac stretch. The review details how alternative splicing of titin pre-mRNA produces different isoforms that greatly impact passive tension and cardiac function, under physiological and pathological conditions. Key posttranslational modifications, especially phosphorylation, play crucial roles in adjusting titin's stiffness, allowing for rapid adaptation to changing hemodynamic demands. Abnormal titin modifications and dysregulation of isoforms are linked to cardiac diseases such as heart failure with preserved ejection fraction, where increased stiffness impairs diastolic function. In addition, the review discusses the importance of the A-band region of titin in setting thick filament length and enhancing Ca²+ sensitivity, contributing to the Frank-Starling Mechanism of the heart. TTN truncating variants are frequently associated with dilated cardiomyopathy, and the review outlines potential disease mechanisms, including haploinsufficiency, sarcomere disarray, and altered thick filament regulation. Variants in TTN have also been linked to conditions such as peripartum cardiomyopathy and chemotherapy-induced cardiomyopathy. Therapeutic avenues are explored, including targeting splicing factors such as RBM20 (RNA binding motif protein 20) to adjust isoform ratios or using engineered heart tissues to study disease mechanisms. Advances in genetic engineering, including CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats), offer promise for modifying TTN to treat titin-related cardiomyopathies. This comprehensive review highlights titin's structural, mechanical, and signaling roles in heart function and the impact of TTN mutations on cardiac diseases.
Collapse
Affiliation(s)
- Henk L Granzier
- Department of Cellular and Molecular Medicine, Molecular Cardiovascular Research Program, The University of Arizona, Tucson (H.L.G.)
| | - Siegfried Labeit
- Department of Integrative Pathophysiology, Medical Faculty Mannheim, DZHK Partnersite Mannheim-Heidelberg, University of Heidelberg, Germany (S.L.)
| |
Collapse
|
3
|
Zhou X, Ding Y, Yang C, Li C, Su Z, Xu J, Qu C, Shi Y, Kang X. FHL3 gene regulates bovine skeletal muscle cell growth through the PI3K/Akt/mTOR signaling pathway. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101356. [PMID: 39549419 DOI: 10.1016/j.cbd.2024.101356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
Beef quality is a critical factor in evaluating the effectiveness of beef cattle production. Fiber types play key roles in determining muscle growth and meat quality characteristics. FHL3 is de novo expressed in skeletal muscle and is responsible for MyHC isoform expression in C2C12 cells. Nevertheless, the precise function of this factor in regulating the proliferation, differentiation, and fiber type of bovine skeletal muscle cells (BSMCs) have yet to be identified. This study aimed to investigate the impact of the FHL3 on BSMCs proliferation, differentiation, and muscle fiber types. The results revealed that the FHL3 promoted BSMCs proliferation, inhibited differentiation, increased type II muscle fiber expression, and decreased type I muscle fiber expression. Meanwhile, the FHL3 promoted the expression and phosphorylation levels of PI3K, Akt, and mTOR in the PI3K/Akt/mTOR signaling pathway, and inhibited the expression and phosphorylation levels of PI3K, Akt, and mTOR after treatment with the pathway inhibitor LY294002, furthermore, it promoted differentiation and inhibited proliferation of BSMCs, while promoting the expression of type II muscle fibers and inhibiting the expression of type I muscle fibers. The results suggest that the FHL3 has an effect on promoting the proliferation and inhibiting the differentiation of BSMCs through the PI3K/Akt/mTOR signaling pathway, but the effect of the FHL3 on myofiber type conversion is not regulated by this pathway. The objective of this study is to enhance our understanding of the molecular function of FHL3 in the development of BSMCs.
Collapse
Affiliation(s)
- Xiaonan Zhou
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Yanling Ding
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Chaoyun Yang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Chenglong Li
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Zonghua Su
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Junjie Xu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Chang Qu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Yuangang Shi
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Xiaolong Kang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China.
| |
Collapse
|
4
|
Jiang X, Ly OT, Chen H, Zhang Z, Ibarra BA, Pavel MA, Brown GE, Sridhar A, Tofovic D, Swick A, Marszalek R, Vanoye CG, Navales F, George AL, Khetani SR, Rehman J, Gao Y, Darbar D, Saxena A. Transient titin-dependent ventricular defects during development lead to adult atrial arrhythmia and impaired contractility. iScience 2024; 27:110395. [PMID: 39100923 PMCID: PMC11296057 DOI: 10.1016/j.isci.2024.110395] [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: 03/14/2024] [Revised: 05/28/2024] [Accepted: 06/25/2024] [Indexed: 08/06/2024] Open
Abstract
Developmental causes of the most common arrhythmia, atrial fibrillation (AF), are poorly defined, with compensation potentially masking arrhythmic risk. Here, we delete 9 amino acids (Δ9) within a conserved domain of the giant protein titin's A-band in zebrafish and human-induced pluripotent stem cell-derived atrial cardiomyocytes (hiPSC-aCMs). We find that ttna Δ9/Δ9 zebrafish embryos' cardiac morphology is perturbed and accompanied by reduced functional output, but ventricular function recovers within days. Despite normal ventricular function, ttna Δ9/Δ9 adults exhibit AF and atrial myopathy, which are recapitulated in TTN Δ9/Δ9-hiPSC-aCMs. Additionally, action potential is shortened and slow delayed rectifier potassium current (I Ks) is increased due to aberrant atrial natriuretic peptide (ANP) levels. Strikingly, suppression of I Ks in both models prevents AF and improves atrial contractility. Thus, a small internal deletion in titin causes developmental abnormalities that increase the risk of AF via ion channel remodeling, with implications for patients who harbor disease-causing variants in sarcomeric proteins.
Collapse
Affiliation(s)
- Xinghang Jiang
- Department of Cell, Developmental, and Integrative Biology, UAB Heersink School of Medicine, Birmingham, AL 35233, USA
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Olivia T. Ly
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Hanna Chen
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ziwei Zhang
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Beatriz A. Ibarra
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Mahmud A. Pavel
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Grace E. Brown
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Arvind Sridhar
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Physiology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - David Tofovic
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Medicine, Jesse Brown Veterans Administration, Chicago, IL 60612, USA
| | - Abigail Swick
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Richard Marszalek
- Department of Physiology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Carlos G. Vanoye
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Fritz Navales
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Alfred L. George
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Salman R. Khetani
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Jalees Rehman
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Yu Gao
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Dawood Darbar
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Medicine, Jesse Brown Veterans Administration, Chicago, IL 60612, USA
| | - Ankur Saxena
- Department of Cell, Developmental, and Integrative Biology, UAB Heersink School of Medicine, Birmingham, AL 35233, USA
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
- O'Neal Comprehensive Cancer Center, Birmingham, AL 35233, USA
| |
Collapse
|
5
|
Strom J, Bull M, Gohlke J, Saripalli C, Methawasin M, Gotthardt M, Granzier H. Titin's cardiac-specific N2B element is critical to mechanotransduction during volume overload of the heart. J Mol Cell Cardiol 2024; 191:40-49. [PMID: 38604403 PMCID: PMC11229416 DOI: 10.1016/j.yjmcc.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 03/09/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
The heart has the ability to detect and respond to changes in mechanical load through a process called mechanotransduction. In this study, we focused on investigating the role of the cardiac-specific N2B element within the spring region of titin, which has been proposed to function as a mechanosensor. To assess its significance, we conducted experiments using N2B knockout (KO) mice and wildtype (WT) mice, subjecting them to three different conditions: 1) cardiac pressure overload induced by transverse aortic constriction (TAC), 2) volume overload caused by aortocaval fistula (ACF), and 3) exercise-induced hypertrophy through swimming. Under conditions of pressure overload (TAC), both genotypes exhibited similar hypertrophic responses. In contrast, WT mice displayed robust left ventricular hypertrophy after one week of volume overload (ACF), while the KO mice failed to undergo hypertrophy and experienced a high mortality rate. Similarly, swim exercise-induced hypertrophy was significantly reduced in the KO mice. RNA-Seq analysis revealed an abnormal β-adrenergic response to volume overload in the KO mice, as well as a diminished response to isoproterenol-induced hypertrophy. Because it is known that the N2B element interacts with the four-and-a-half LIM domains 1 and 2 (FHL1 and FHL2) proteins, both of which have been associated with mechanotransduction, we evaluated these proteins. Interestingly, while volume-overload resulted in FHL1 protein expression levels that were comparable between KO and WT mice, FHL2 protein levels were reduced by over 90% in the KO mice compared to WT. This suggests that in response to volume overload, FHL2 might act as a signaling mediator between the N2B element and downstream signaling pathways. Overall, our study highlights the importance of the N2B element in mechanosensing during volume overload, both in physiological and pathological settings.
Collapse
Affiliation(s)
- Joshua Strom
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, United States of America; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, United States of America
| | - Mathew Bull
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, United States of America; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, United States of America
| | - Jochen Gohlke
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, United States of America; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, United States of America
| | - Chandra Saripalli
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, United States of America; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, United States of America
| | - Mei Methawasin
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, United States of America; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, United States of America
| | - Michael Gotthardt
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, United States of America.
| |
Collapse
|
6
|
Sun Y, Liu X, Huang W, Le S, Yan J. Structural domain in the Titin N2B-us region binds to FHL2 in a force-activation dependent manner. Nat Commun 2024; 15:4496. [PMID: 38802383 PMCID: PMC11530556 DOI: 10.1038/s41467-024-48828-7] [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: 03/31/2022] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Titin N2B unique sequence (N2B-us) is a 572 amino acid sequence that acts as an elastic spring to regulate muscle passive elasticity. It is thought to lack stable tertiary structures and is a force-bearing region that is regulated by mechanical stretching. In this study, the conformation of N2B-us and its interaction with four-and-a-half LIM domain protein 2 (FHL2) are investigated using AlphaFold2 predictions and single-molecule experimental validation. Surprisingly, a stable alpha/beta structural domain is predicted and confirmed in N2B-us that can be mechanically unfolded at forces of a few piconewtons. Additionally, more than twenty FHL2 LIM domain binding sites are predicted to spread throughout N2B-us. Single-molecule manipulation experiments reveals the force-dependent binding of FHL2 to the N2B-us structural domain. These findings provide insights into the mechano-sensing functions of N2B-us and its interactions with FHL2.
Collapse
Affiliation(s)
- Yuze Sun
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Xuyao Liu
- Department of Physics, National University of Singapore, Singapore, Singapore
| | - Wenmao Huang
- Department of Physics, National University of Singapore, Singapore, Singapore
| | - Shimin Le
- Department of Physics, National University of Singapore, Singapore, Singapore
| | - Jie Yan
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
- Department of Physics, National University of Singapore, Singapore, Singapore.
- Centre for Biological Imaging Sciences, National University of Singapore, Singapore, Singapore.
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, China.
| |
Collapse
|
7
|
Li C, Warren DT, Zhou C, De Silva S, Wilson DGS, Garcia-Maya M, Wheeler MA, Meinke P, Sawyer G, Ehler E, Wehnert M, Rao L, Zhang Q, Shanahan CM. Nesprin-2 is a novel scaffold protein for telethonin and FHL-2 in the cardiomyocyte sarcomere. J Biol Chem 2024; 300:107254. [PMID: 38569934 PMCID: PMC11078644 DOI: 10.1016/j.jbc.2024.107254] [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: 09/05/2023] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
Nesprins comprise a family of multi-isomeric scaffolding proteins, forming the linker of nucleoskeleton-and-cytoskeleton complex with lamin A/C, emerin and SUN1/2 at the nuclear envelope. Mutations in nesprin-1/-2 are associated with Emery-Dreifuss muscular dystrophy (EDMD) with conduction defects and dilated cardiomyopathy (DCM). We have previously observed sarcomeric staining of nesprin-1/-2 in cardiac and skeletal muscle, but nesprin function in this compartment remains unknown. In this study, we show that specific nesprin-2 isoforms are highly expressed in cardiac muscle and localize to the Z-disc and I band of the sarcomere. Expression of GFP-tagged nesprin-2 giant spectrin repeats 52 to 53, localized to the sarcomere of neonatal rat cardiomyocytes. Yeast two-hybrid screening of a cardiac muscle cDNA library identified telethonin and four-and-half LIM domain (FHL)-2 as potential nesprin-2 binding partners. GST pull-down and immunoprecipitation confirmed the individual interactions between nesprin-2/telethonin and nesprin-2/FHL-2, and showed that nesprin-2 and telethonin binding was dependent on telethonin phosphorylation status. Importantly, the interactions between these binding partners were impaired by mutations in nesprin-2, telethonin, and FHL-2 identified in EDMD with DCM and hypertrophic cardiomyopathy patients. These data suggest that nesprin-2 is a novel sarcomeric scaffold protein that may potentially participate in the maintenance and/or regulation of sarcomeric organization and function.
Collapse
Affiliation(s)
- Chen Li
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK; Department of Cardiology, West China Hospital of Sichuan University, Chengdu, China
| | - Derek T Warren
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK; School of Pharmacy, University of East Anglia, Norwich, UK
| | - Can Zhou
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK
| | - Shanelle De Silva
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK
| | - Darren G S Wilson
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK
| | - Mitla Garcia-Maya
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, London, UK
| | - Matthew A Wheeler
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Peter Meinke
- Friedrich-Baur-Institute at the Department of Neurology, LMU University Hospital, Munich, Germany
| | - Greta Sawyer
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK
| | - Elisabeth Ehler
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK; Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, London, UK
| | - Manfred Wehnert
- Institute of Human Genetics, University of Greifswald, Greifswald, Germany
| | - Li Rao
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, China
| | - Qiuping Zhang
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK.
| | - Catherine M Shanahan
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK.
| |
Collapse
|
8
|
Chatterjee S, Leach-Mehrwald M, Huang CK, Xiao K, Fuchs M, Otto M, Lu D, Dang V, Winkler T, Dunbar CE, Thum T, Bär C. Telomerase is essential for cardiac differentiation and sustained metabolism of human cardiomyocytes. Cell Mol Life Sci 2024; 81:196. [PMID: 38658440 PMCID: PMC11043037 DOI: 10.1007/s00018-024-05239-7] [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: 10/24/2023] [Revised: 03/29/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
Telomeres as the protective ends of linear chromosomes, are synthesized by the enzyme telomerase (TERT). Critically short telomeres essentially contribute to aging-related diseases and are associated with a broad spectrum of disorders known as telomeropathies. In cardiomyocytes, telomere length is strongly correlated with cardiomyopathies but it remains ambiguous whether short telomeres are the cause or the result of the disease. In this study, we employed an inducible CRISPRi human induced pluripotent stem cell (hiPSC) line to silence TERT expression enabling the generation of hiPSCs and hiPSC-derived cardiomyocytes with long and short telomeres. Reduced telomerase activity and shorter telomere lengths of hiPSCs induced global transcriptomic changes associated with cardiac developmental pathways. Consequently, the differentiation potential towards cardiomyocytes was strongly impaired and single cell RNA sequencing revealed a shift towards a more smooth muscle cell like identity in the cells with the shortest telomeres. Poor cardiomyocyte function and increased sensitivity to stress directly correlated with the extent of telomere shortening. Collectively our data demonstrates a TERT dependent cardiomyogenic differentiation defect, highlighting the CRISPRi TERT hiPSCs model as a powerful platform to study the mechanisms and consequences of short telomeres in the heart and also in the context of telomeropathies.
Collapse
Affiliation(s)
- Shambhabi Chatterjee
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- Center of Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Megan Leach-Mehrwald
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Cheng-Kai Huang
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Ke Xiao
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Maximilian Fuchs
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Mandy Otto
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Dongchao Lu
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- Center of Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Vinh Dang
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Winkler
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- Center of Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.
- Center of Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany.
| |
Collapse
|
9
|
Stroik D, Gregorich ZR, Raza F, Ge Y, Guo W. Titin: roles in cardiac function and diseases. Front Physiol 2024; 15:1385821. [PMID: 38660537 PMCID: PMC11040099 DOI: 10.3389/fphys.2024.1385821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The giant protein titin is an essential component of muscle sarcomeres. A single titin molecule spans half a sarcomere and mediates diverse functions along its length by virtue of its unique domains. The A-band of titin functions as a molecular blueprint that defines the length of the thick filaments, the I-band constitutes a molecular spring that determines cell-based passive stiffness, and various domains, including the Z-disk, I-band, and M-line, serve as scaffolds for stretch-sensing signaling pathways that mediate mechanotransduction. This review aims to discuss recent insights into titin's functional roles and their relationship to cardiac function. The role of titin in heart diseases, such as dilated cardiomyopathy and heart failure with preserved ejection fraction, as well as its potential as a therapeutic target, is also discussed.
Collapse
Affiliation(s)
- Dawson Stroik
- Cellular and Molecular Pathology Program, Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Department of Animal and Dairy Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI, United States
| | - Zachery R. Gregorich
- Department of Animal and Dairy Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI, United States
| | - Farhan Raza
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ying Ge
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Wei Guo
- Cellular and Molecular Pathology Program, Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Department of Animal and Dairy Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
10
|
Dennhag N, Kahsay A, Nissen I, Nord H, Chermenina M, Liu J, Arner A, Liu JX, Backman LJ, Remeseiro S, von Hofsten J, Pedrosa Domellöf F. fhl2b mediates extraocular muscle protection in zebrafish models of muscular dystrophies and its ectopic expression ameliorates affected body muscles. Nat Commun 2024; 15:1950. [PMID: 38431640 PMCID: PMC10908798 DOI: 10.1038/s41467-024-46187-x] [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: 09/12/2023] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
In muscular dystrophies, muscle fibers loose integrity and die, causing significant suffering and premature death. Strikingly, the extraocular muscles (EOMs) are spared, functioning well despite the disease progression. Although EOMs have been shown to differ from body musculature, the mechanisms underlying this inherent resistance to muscle dystrophies remain unknown. Here, we demonstrate important differences in gene expression as a response to muscle dystrophies between the EOMs and trunk muscles in zebrafish via transcriptomic profiling. We show that the LIM-protein Fhl2 is increased in response to the knockout of desmin, plectin and obscurin, cytoskeletal proteins whose knockout causes different muscle dystrophies, and contributes to disease protection of the EOMs. Moreover, we show that ectopic expression of fhl2b can partially rescue the muscle phenotype in the zebrafish Duchenne muscular dystrophy model sapje, significantly improving their survival. Therefore, Fhl2 is a protective agent and a candidate target gene for therapy of muscular dystrophies.
Collapse
Affiliation(s)
- Nils Dennhag
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - Abraha Kahsay
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - Itzel Nissen
- Department of Medical and Translational Biology; Section of Molecular Medicine, Umeå University, Umeå, Sweden
- Wallenberg Center for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Hanna Nord
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Maria Chermenina
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - Jiao Liu
- Div. Thoracic Surgery, Dept. Clinical Sciences, Lund University, Lund, Sweden
- College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Anders Arner
- Div. Thoracic Surgery, Dept. Clinical Sciences, Lund University, Lund, Sweden
| | - Jing-Xia Liu
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Ludvig J Backman
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Silvia Remeseiro
- Department of Medical and Translational Biology; Section of Molecular Medicine, Umeå University, Umeå, Sweden
- Wallenberg Center for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Jonas von Hofsten
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden.
| | - Fatima Pedrosa Domellöf
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden.
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden.
| |
Collapse
|
11
|
Qi T, Zhang J, Zhang K, Zhang W, Song Y, Lian K, Kan C, Han F, Hou N, Sun X. Unraveling the role of the FHL family in cardiac diseases: Mechanisms, implications, and future directions. Biochem Biophys Res Commun 2024; 694:149468. [PMID: 38183876 DOI: 10.1016/j.bbrc.2024.149468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
Heart diseases are a major cause of morbidity and mortality worldwide. Understanding the molecular mechanisms underlying these diseases is essential for the development of effective diagnostic and therapeutic strategies. The FHL family consists of five members: FHL1, FHL2, FHL3, FHL4, and FHL5/Act. These members exhibit different expression patterns in various tissues including the heart. FHL family proteins are implicated in cardiac remodeling, regulation of metabolic enzymes, and cardiac biomechanical stress perception. A large number of studies have explored the link between FHL family proteins and cardiac disease, skeletal muscle disease, and ovarian metabolism, but a comprehensive and in-depth understanding of the specific molecular mechanisms targeting FHL on cardiac disease is lacking. The aim of this review is to explore the structure and function of FHL family members, to comprehensively elucidate the mechanisms by which they regulate the heart, and to explore in depth the changes in FHL family members observed in different cardiac disorders, as well as the effects of mutations in FHL proteins on heart health.
Collapse
Affiliation(s)
- Tongbing Qi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Wenqiang Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Yixin Song
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Kexin Lian
- Department of Nephrology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| |
Collapse
|
12
|
Tojo T, Yamaoka-Tojo M. Molecular Mechanisms Underlying the Progression of Aortic Valve Stenosis: Bioinformatic Analysis of Signal Pathways and Hub Genes. Int J Mol Sci 2023; 24:ijms24097964. [PMID: 37175670 PMCID: PMC10177913 DOI: 10.3390/ijms24097964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
The calcification of the aortic valve causes increased leaflet stiffness and leads to the development and progression of stenotic aortic valve disease. However, the molecular and cellular mechanisms underlying stenotic calcification remain poorly understood. Herein, we examined the gene expression associated with valve calcification and the progression of calcific aortic valve stenosis. We downloaded two publicly available gene expression profiles (GSE83453 and GSE51472) from NCBI-Gene Expression Omnibus database for the combined analysis of samples from human aortic stenosis and normal aortic valve tissue. After identifying the differentially expressed genes (DEGs) using the GEO2R online tool, we performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses. We also analyzed the protein-protein interactions (PPIs) of the DEGs using the NetworkAnalyst online tool. We identified 4603 upregulated and 6272 downregulated DEGs, which were enriched in the positive regulation of cell adhesion, leukocyte-mediated immunity, response to hormones, cytokine signaling in the immune system, lymphocyte activation, and growth hormone receptor signaling. PPI network analysis identified 10 hub genes: VCAM1, FHL2, RUNX1, TNFSF10, PLAU, SPOCK1, CD74, SIPA1L2, TRIB1, and CXCL12. Through bioinformatic analysis, we identified potential biomarkers and therapeutic targets for aortic stenosis, providing a theoretical basis for future studies.
Collapse
Affiliation(s)
- Taiki Tojo
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Minako Yamaoka-Tojo
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
- Department of Rehabilitation, Kitasato University School of Allied Health Sciences, Sagamihara 252-0373, Japan
| |
Collapse
|
13
|
Grogan A, Huang W, Brong A, Kane MA, Kontrogianni-Konstantopoulos A. Alterations in cytoskeletal and Ca 2+ cycling regulators in atria lacking the obscurin Ig58/59 module. Front Cardiovasc Med 2023; 10:1085840. [PMID: 37304957 PMCID: PMC10251194 DOI: 10.3389/fcvm.2023.1085840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/26/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Obscurin (720-870 kDa) is a giant cytoskeletal and signaling protein that possesses both structural and regulatory functions in striated muscles. Immunoglobulin domains 58/59 (Ig58/59) of obscurin bind to a diverse set of proteins that are essential for the proper structure and function of the heart, including giant titin, novex-3, and phospholamban (PLN). Importantly, the pathophysiological significance of the Ig58/59 module has been further underscored by the discovery of several mutations within Ig58/59 that are linked to various forms of myopathy in humans. We previously generated a constitutive deletion mouse model, Obscn-ΔIg58/59, that expresses obscurin lacking Ig58/59, and characterized the effects of this deletion on cardiac morphology and function through aging. Our findings demonstrated that Obscn-ΔIg58/59 male animals develop severe arrhythmia, primarily manifesting as episodes of junctional escape and spontaneous loss of regular p-waves, reminiscent of human atrial fibrillation, accompanied by significant atrial enlargement that progresses in severity with aging. Methods and Results To comprehensively characterize the molecular alterations responsible for these pathologies, we performed proteomic and phospho-proteomic analyses in aging Obscn-ΔIg58/59 atria. Our studies revealed extensive and novel alterations in the expression and phosphorylation profile of major cytoskeletal proteins, Ca2+ regulators, and Z-disk associated protein complexes in the Obscn-ΔIg58/59 atria through aging. Discussion These studies implicate obscurin, particularly the Ig58/59 module, as an essential regulator of the Z-disk associated cytoskeleton and Ca2+ cycling in the atria and provide new molecular insights into the development of atrial fibrillation and remodeling.
Collapse
Affiliation(s)
- Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, United States
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Annie Brong
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, United States
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | | |
Collapse
|
14
|
McNamara JW, Parker BL, Voges HK, Mehdiabadi NR, Bolk F, Ahmad F, Chung JD, Charitakis N, Molendijk J, Zech ATL, Lal S, Ramialison M, Karavendzas K, Pointer HL, Syrris P, Lopes LR, Elliott PM, Lynch GS, Mills RJ, Hudson JE, Watt KI, Porrello ER, Elliott DA. Alpha kinase 3 signaling at the M-band maintains sarcomere integrity and proteostasis in striated muscle. NATURE CARDIOVASCULAR RESEARCH 2023; 2:159-173. [PMID: 39196058 PMCID: PMC11358020 DOI: 10.1038/s44161-023-00219-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/19/2023] [Indexed: 08/29/2024]
Abstract
Muscle contraction is driven by the molecular machinery of the sarcomere. As phosphorylation is a critical regulator of muscle function, the identification of regulatory kinases is important for understanding sarcomere biology. Pathogenic variants in alpha kinase 3 (ALPK3) cause cardiomyopathy and musculoskeletal disease, but little is known about this atypical kinase. Here we show that ALPK3 is an essential component of the M-band of the sarcomere and define the ALPK3-dependent phosphoproteome. ALPK3 deficiency impaired contractility both in human cardiac organoids and in the hearts of mice harboring a pathogenic truncating Alpk3 variant. ALPK3-dependent phosphopeptides were enriched for sarcomeric components of the M-band and the ubiquitin-binding protein sequestosome-1 (SQSTM1) (also known as p62). Analysis of the ALPK3 interactome confirmed binding to M-band proteins including SQSTM1. In human pluripotent stem cell-derived cardiomyocytes modeling cardiomyopathic ALPK3 mutations, sarcomeric organization and M-band localization of SQSTM1 were abnormal suggesting that this mechanism may underly disease pathogenesis.
Collapse
Affiliation(s)
- James W McNamara
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Benjamin L Parker
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Holly K Voges
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Neda R Mehdiabadi
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Francesca Bolk
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Feroz Ahmad
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Jin D Chung
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Natalie Charitakis
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Jeffrey Molendijk
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Antonia T L Zech
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Sean Lal
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia
| | - Mirana Ramialison
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Kathy Karavendzas
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Hayley L Pointer
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Petros Syrris
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, UK
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Luis R Lopes
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, UK
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Perry M Elliott
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, UK
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Gordon S Lynch
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Richard J Mills
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - James E Hudson
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kevin I Watt
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Enzo R Porrello
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia.
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia.
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
| | - David A Elliott
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
15
|
Meyer DJ, Díaz-García CM, Nathwani N, Rahman M, Yellen G. The Na +/K + pump dominates control of glycolysis in hippocampal dentate granule cells. eLife 2022; 11:e81645. [PMID: 36222651 PMCID: PMC9592084 DOI: 10.7554/elife.81645] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular ATP that is consumed to perform energetically expensive tasks must be replenished by new ATP through the activation of metabolism. Neuronal stimulation, an energetically demanding process, transiently activates aerobic glycolysis, but the precise mechanism underlying this glycolysis activation has not been determined. We previously showed that neuronal glycolysis is correlated with Ca2+ influx, but is not activated by feedforward Ca2+ signaling (Díaz-García et al., 2021a). Since ATP-powered Na+ and Ca2+ pumping activities are increased following stimulation to restore ion gradients and are estimated to consume most neuronal ATP, we aimed to determine if they are coupled to neuronal glycolysis activation. By using two-photon imaging of fluorescent biosensors and dyes in dentate granule cell somas of acute mouse hippocampal slices, we observed that production of cytoplasmic NADH, a byproduct of glycolysis, is strongly coupled to changes in intracellular Na+, while intracellular Ca2+ could only increase NADH production if both forward Na+/Ca2+ exchange and Na+/K+ pump activity were intact. Additionally, antidromic stimulation-induced intracellular [Na+] increases were reduced >50% by blocking Ca2+ entry. These results indicate that neuronal glycolysis activation is predominantly a response to an increase in activity of the Na+/K+ pump, which is strongly potentiated by Na+ influx through the Na+/Ca2+ exchanger during extrusion of Ca2+ following stimulation.
Collapse
Affiliation(s)
- Dylan J Meyer
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | | | - Nidhi Nathwani
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Mahia Rahman
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Gary Yellen
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
16
|
Stathopoulou K, Schnittger J, Raabe J, Fleischer F, Mangels N, Piasecki A, Findlay J, Hartmann K, Krasemann S, Schlossarek S, Uebeler J, Wixler V, Blake DJ, Baillie GS, Carrier L, Ehler E, Cuello F. CMYA5 is a novel interaction partner of FHL2 in cardiac myocytes. FEBS J 2022; 289:4622-4645. [PMID: 35176204 DOI: 10.1111/febs.16402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 01/13/2022] [Accepted: 02/15/2022] [Indexed: 11/27/2022]
Abstract
Four-and-a-half LIM domains protein 2 (FHL2) is an anti-hypertrophic adaptor protein that regulates cardiac myocyte signalling and function. Herein, we identified cardiomyopathy-associated 5 (CMYA5) as a novel FHL2 interaction partner in cardiac myocytes. In vitro pull-down assays demonstrated interaction between FHL2 and the N- and C-terminal regions of CMYA5. The interaction was verified in adult cardiac myocytes by proximity ligation assays. Immunofluorescence and confocal microscopy demonstrated co-localisation in the same subcellular compartment. The binding interface between FHL2 and CMYA5 was mapped by peptide arrays. Exposure of neonatal rat ventricular myocytes to a CMYA5 peptide covering one of the FHL2 interaction sites led to an increase in cell area at baseline, but a blunted response to chronic phenylephrine treatment. In contrast to wild-type hearts, loss or reduced FHL2 expression in Fhl2-targeted knockout mouse hearts or in a humanised mouse model of hypertrophic cardiomyopathy led to redistribution of CMYA5 into the perinuclear and intercalated disc region. Taken together, our results indicate a direct interaction of the two adaptor proteins FHL2 and CMYA5 in cardiac myocytes, which might impact subcellular compartmentation of CMYA5.
Collapse
Affiliation(s)
- Konstantina Stathopoulou
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Josef Schnittger
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Janice Raabe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Frederic Fleischer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Nils Mangels
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Germany
| | - Angelika Piasecki
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Jane Findlay
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Kristin Hartmann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Saskia Schlossarek
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - June Uebeler
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Viktor Wixler
- Institute of Molecular Virology, Centre for Molecular Biology of Inflammation, Westfaelische Wilhelms-University, Germany
| | - Derek J Blake
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - George S Baillie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Elisabeth Ehler
- School of Cardiovascular Medicine and Sciences, BHF Research Excellence Centre, King's College London, UK.,Randall Centre for Cell and Molecular Biophysics (School of Basic and Medical Biosciences), King's College London, UK
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| |
Collapse
|
17
|
Lamber EP, Guicheney P, Pinotsis N. The role of the M-band myomesin proteins in muscle integrity and cardiac disease. J Biomed Sci 2022; 29:18. [PMID: 35255917 PMCID: PMC8900313 DOI: 10.1186/s12929-022-00801-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
Transversal structural elements in cross-striated muscles, such as the M-band or the Z-disc, anchor and mechanically stabilize the contractile apparatus and its minimal unit—the sarcomere. The ability of proteins to target and interact with these structural sarcomeric elements is an inevitable necessity for the correct assembly and functionality of the myofibrillar apparatus. Specifically, the M-band is a well-recognized mechanical and signaling hub dealing with active forces during contraction, while impairment of its function leads to disease and death. Research on the M-band architecture is focusing on the assembly and interactions of the three major filamentous proteins in the region, mainly the three myomesin proteins including their embryonic heart (EH) isoform, titin and obscurin. These proteins form the basic filamentous network of the M-band, interacting with each other as also with additional proteins in the region that are involved in signaling, energetic or mechanosensitive processes. While myomesin-1, titin and obscurin are found in every muscle, the expression levels of myomesin-2 (also known as M-protein) and myomesin-3 are tissue specific: myomesin-2 is mainly expressed in the cardiac and fast skeletal muscles, while myomesin-3 is mainly expressed in intermediate muscles and specific regions of the cardiac muscle. Furthermore, EH-myomesin apart from its role during embryonic stages, is present in adults with specific cardiac diseases. The current work in structural, molecular, and cellular biology as well as in animal models, provides important details about the assembly of myomesin-1, obscurin and titin, the information however about the myomesin-2 and -3, such as their interactions, localization and structural details remain very limited. Remarkably, an increasing number of reports is linking all three myomesin proteins and particularly myomesin-2 to serious cardiovascular diseases suggesting that this protein family could be more important than originally thought. In this review we will focus on the myomesin protein family, the myomesin interactions and structural differences between isoforms and we will provide the most recent evidence why the structurally and biophysically unexplored myomesin-2 and myomesin-3 are emerging as hot targets for understanding muscle function and disease.
Collapse
|
18
|
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: 21] [Impact Index Per Article: 7.0] [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
|
19
|
Su K, Mayans O, Diederichs K, Fleming JR. Pairwise sequence similarity mapping with PaSiMap: Reclassification of immunoglobulin domains from titin as case study. Comput Struct Biotechnol J 2022; 20:5409-5419. [PMID: 36212532 PMCID: PMC9529554 DOI: 10.1016/j.csbj.2022.09.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
A novel multidimensional scaling pipeline for sequence analysis. A simple way to distinguish between unique and shared sequence features. Titin domains were reclassified, improving upon earlier analysis.
Sequence comparison is critical for the functional assignment of newly identified protein genes. As uncharacterized protein sequences accumulate, there is an increasing need for sensitive tools for their classification. Here, we present a novel multidimensional scaling pipeline, PaSiMap, which creates a map of pairwise sequence similarities. Uniquely, PaSiMap distinguishes between unique and shared features, allowing for a distinct view of protein-sequence relationships. We demonstrate PaSiMap’s efficiency in detecting sequence groups and outliers using titin’s 169 immunoglobulin (Ig) domains. We show that Ig domain similarity is hierarchical, being firstly determined by chain location, then by the loop features of the Ig fold and, finally, by super-repeat position. The existence of a previously unidentified domain repeat in the distal, constitutive I-band is revealed. Prototypic Igs, plus notable outliers, are identified and thereby domain classification improved. This re-classification can now guide future molecular research. In summary, we demonstrate that PaSiMap is a sensitive tool for the classification of protein sequences, which adds a new perspective in the understanding of inter-protein relationships. PaSiMap is applicable to any biological system defined by a linear sequence, including polynucleotide chains.
Collapse
|
20
|
Radke MH, Badillo-Lisakowski V, Britto-Borges T, Kubli DA, Jüttner R, Parakkat P, Carballo JL, Hüttemeister J, Liss M, Hansen A, Dieterich C, Mullick AE, Gotthardt M. Therapeutic inhibition of RBM20 improves diastolic function in a murine heart failure model and human engineered heart tissue. Sci Transl Med 2021; 13:eabe8952. [PMID: 34851694 DOI: 10.1126/scitranslmed.abe8952] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Michael H Radke
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany
| | - Victor Badillo-Lisakowski
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany.,Humboldt-Universität zu Berlin, 10099 Berlin, Germany
| | - Thiago Britto-Borges
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology and Department of Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | | | - René Jüttner
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Pragati Parakkat
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany.,Department of Cardiology, Charité Universitätsmedizin Berlin, 10115 Berlin, Germany
| | - Jacobo Lopez Carballo
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,Department of Cardiology, Charité Universitätsmedizin Berlin, 10115 Berlin, Germany
| | - Judith Hüttemeister
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany
| | - Martin Liss
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany
| | - Arne Hansen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology and Department of Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | | | - Michael Gotthardt
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany.,Department of Cardiology, Charité Universitätsmedizin Berlin, 10115 Berlin, Germany
| |
Collapse
|
21
|
Biophysical Reviews "Meet the Editor Series"-Elisabeth Ehler. Biophys Rev 2021; 13:579-581. [PMID: 34777613 DOI: 10.1007/s12551-021-00830-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 08/14/2021] [Indexed: 10/20/2022] Open
|
22
|
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: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
23
|
Solís C, Russell B. Striated muscle proteins are regulated both by mechanical deformation and by chemical post-translational modification. Biophys Rev 2021; 13:679-695. [PMID: 34777614 PMCID: PMC8555064 DOI: 10.1007/s12551-021-00835-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/23/2021] [Indexed: 01/09/2023] Open
Abstract
All cells sense force and build their cytoskeleton to optimize function. How is this achieved? Two major systems are involved. The first is that load deforms specific protein structures in a proportional and orientation-dependent manner. The second is post-translational modification of proteins as a consequence of signaling pathway activation. These two processes work together in a complex way so that local subcellular assembly as well as overall cell function are controlled. This review discusses many cell types but focuses on striated muscle. Detailed information is provided on how load deforms the structure of proteins in the focal adhesions and filaments, using α-actinin, vinculin, talin, focal adhesion kinase, LIM domain-containing proteins, filamin, myosin, titin, and telethonin as examples. Second messenger signals arising from external triggers are distributed throughout the cell causing post-translational or chemical modifications of protein structures, with the actin capping protein CapZ and troponin as examples. There are numerous unanswered questions of how mechanical and chemical signals are integrated by muscle proteins to regulate sarcomere structure and function yet to be studied. Therefore, more research is needed to see how external triggers are integrated with local tension generated within the cell. Nonetheless, maintenance of tension in the sarcomere is the essential and dominant mechanism, leading to the well-known phrase in exercise physiology: "use it or lose it."
Collapse
Affiliation(s)
- Christopher Solís
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Brenda Russell
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| |
Collapse
|
24
|
Biquand A, Spinozzi S, Tonino P, Cosette J, Strom J, Elbeck Z, Knöll R, Granzier H, Lostal W, Richard I. Titin M-line insertion sequence 7 is required for proper cardiac function in mice. J Cell Sci 2021; 134:271843. [PMID: 34401916 DOI: 10.1242/jcs.258684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 08/06/2021] [Indexed: 11/20/2022] Open
Abstract
Titin is a giant sarcomeric protein that is involved in a large number of functions, with a primary role in skeletal and cardiac sarcomere organization and stiffness. The titin gene (TTN) is subject to various alternative splicing events, but in the region that is present at the M-line, the only exon that can be spliced out is Mex5, which encodes for the insertion sequence 7 (is7). Interestingly, in the heart, the majority of titin isoforms are Mex5+, suggesting a cardiac role for is7. Here, we performed comprehensive functional, histological, transcriptomic, microscopic and molecular analyses of a mouse model lacking the Ttn Mex5 exon (ΔMex5), and revealed that the absence of the is7 is causative for dilated cardiomyopathy. ΔMex5 mice showed altered cardiac function accompanied by increased fibrosis and ultrastructural alterations. Abnormal expression of excitation-contraction coupling proteins was also observed. The results reported here confirm the importance of the C-terminal region of titin in cardiac function and are the first to suggest a possible relationship between the is7 and excitation-contraction coupling. Finally, these findings give important insights for the identification of new targets in the treatment of titinopathies.
Collapse
Affiliation(s)
- Ariane Biquand
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR_S951, 91000 Evry-Courcouronnes, France
| | - Simone Spinozzi
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR_S951, 91000 Evry-Courcouronnes, France
| | - Paola Tonino
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| | | | - Joshua Strom
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| | - Zaher Elbeck
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Heart and Vascular Theme, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Ralph Knöll
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Heart and Vascular Theme, Karolinska Institutet, 141 57 Huddinge, Sweden.,Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| | - William Lostal
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR_S951, 91000 Evry-Courcouronnes, France
| | - Isabelle Richard
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR_S951, 91000 Evry-Courcouronnes, France
| |
Collapse
|
25
|
Solís C, Solaro RJ. Novel insights into sarcomere regulatory systems control of cardiac thin filament activation. J Gen Physiol 2021; 153:211903. [PMID: 33740037 PMCID: PMC7988513 DOI: 10.1085/jgp.202012777] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Our review focuses on sarcomere regulatory mechanisms with a discussion of cardiac-specific modifications to the three-state model of thin filament activation from a blocked to closed to open state. We discuss modulation of these thin filament transitions by Ca2+, by crossbridge interactions, and by thick filament–associated proteins, cardiac myosin–binding protein C (cMyBP-C), cardiac regulatory light chain (cRLC), and titin. Emerging evidence supports the idea that the cooperative activation of the thin filaments despite a single Ca2+ triggering regulatory site on troponin C (cTnC) cannot be considered in isolation of other functional domains of the sarcomere. We discuss long- and short-range interactions among these domains with the regulatory units of thin filaments, including proteins at the barbed end at the Z-disc and the pointed end near the M-band. Important to these discussions is the ever-increasing understanding of the role of cMyBP-C, cRLC, and titin filaments. Detailed knowledge of these control processes is critical to the understanding of mechanisms sustaining physiological cardiac state with varying hemodynamic load, to better defining genetic and acquired cardiac disorders, and to developing targets for therapies at the level of the sarcomeres.
Collapse
Affiliation(s)
- Christopher Solís
- University of Illinois at Chicago, College of Medicine, Department of Physiology and Biophysics and Center for Cardiovascular Research, Chicago, IL
| | - R John Solaro
- University of Illinois at Chicago, College of Medicine, Department of Physiology and Biophysics and Center for Cardiovascular Research, Chicago, IL
| |
Collapse
|
26
|
Fleming JR, Rani A, Kraft J, Zenker S, Börgeson E, Lange S. Exploring Obscurin and SPEG Kinase Biology. J Clin Med 2021; 10:jcm10050984. [PMID: 33801198 PMCID: PMC7957886 DOI: 10.3390/jcm10050984] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 12/15/2022] Open
Abstract
Three members of the obscurin protein family that contain tandem kinase domains with important signaling functions for cardiac and striated muscles are the giant protein obscurin, its obscurin-associated kinase splice isoform, and the striated muscle enriched protein kinase (SPEG). While there is increasing evidence for the specific roles that each individual kinase domain plays in cross-striated muscles, their biology and regulation remains enigmatic. Our present study focuses on kinase domain 1 and the adjacent low sequence complexity inter-kinase domain linker in obscurin and SPEG. Using Phos-tag gels, we show that the linker in obscurin contains several phosphorylation sites, while the same region in SPEG remained unphosphorylated. Our homology modeling, mutational analysis and molecular docking demonstrate that kinase 1 in obscurin harbors all key amino acids important for its catalytic function and that actions of this domain result in autophosphorylation of the protein. Our bioinformatics analyses also assign a list of putative substrates for kinase domain 1 in obscurin and SPEG, based on the known and our newly proposed phosphorylation sites in muscle proteins, including obscurin itself.
Collapse
Affiliation(s)
- Jennifer R. Fleming
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
- Correspondence: (J.R.F.); (E.B.); (S.L.)
| | - Alankrita Rani
- Centre for Molecular and Translational Medicine, The Wallenberg Laboratory and Wallenberg, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden; (A.R.); (J.K.)
| | - Jamie Kraft
- Centre for Molecular and Translational Medicine, The Wallenberg Laboratory and Wallenberg, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden; (A.R.); (J.K.)
| | - Sanja Zenker
- Department of Medicine, University of California, San Diego, CA 92093, USA;
| | - Emma Börgeson
- Centre for Molecular and Translational Medicine, The Wallenberg Laboratory and Wallenberg, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden; (A.R.); (J.K.)
- Department of Clinical Physiology, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
- Correspondence: (J.R.F.); (E.B.); (S.L.)
| | - Stephan Lange
- Centre for Molecular and Translational Medicine, The Wallenberg Laboratory and Wallenberg, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden; (A.R.); (J.K.)
- Department of Medicine, University of California, San Diego, CA 92093, USA;
- Correspondence: (J.R.F.); (E.B.); (S.L.)
| |
Collapse
|
27
|
Rees M, Nikoopour R, Fukuzawa A, Kho AL, Fernandez-Garcia MA, Wraige E, Bodi I, Deshpande C, Özdemir Ö, Daimagüler HS, Pfuhl M, Holt M, Brandmeier B, Grover S, Fluss J, Longman C, Farrugia ME, Matthews E, Hanna M, Muntoni F, Sarkozy A, Phadke R, Quinlivan R, Oates EC, Schröder R, Thiel C, Reimann J, Voermans N, Erasmus C, Kamsteeg EJ, Konersman C, Grosmann C, McKee S, Tirupathi S, Moore SA, Wilichowski E, Hobbiebrunken E, Dekomien G, Richard I, Van den Bergh P, Domínguez-González C, Cirak S, Ferreiro A, Jungbluth H, Gautel M. Making sense of missense variants in TTN-related congenital myopathies. Acta Neuropathol 2021; 141:431-453. [PMID: 33449170 PMCID: PMC7882473 DOI: 10.1007/s00401-020-02257-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/20/2020] [Accepted: 12/20/2020] [Indexed: 12/15/2022]
Abstract
Mutations in the sarcomeric protein titin, encoded by TTN, are emerging as a common cause of myopathies. The diagnosis of a TTN-related myopathy is, however, often not straightforward due to clinico-pathological overlap with other myopathies and the prevalence of TTN variants in control populations. Here, we present a combined clinico-pathological, genetic and biophysical approach to the diagnosis of TTN-related myopathies and the pathogenicity ascertainment of TTN missense variants. We identified 30 patients with a primary TTN-related congenital myopathy (CM) and two truncating variants, or one truncating and one missense TTN variant, or homozygous for one TTN missense variant. We found that TTN-related myopathies show considerable overlap with other myopathies but are strongly suggested by a combination of certain clinico-pathological features. Presentation was typically at birth with the clinical course characterized by variable progression of weakness, contractures, scoliosis and respiratory symptoms but sparing of extraocular muscles. Cardiac involvement depended on the variant position. Our biophysical analyses demonstrated that missense mutations associated with CMs are strongly destabilizing and exert their effect when expressed on a truncating background or in homozygosity. We hypothesise that destabilizing TTN missense mutations phenocopy truncating variants and are a key pathogenic feature of recessive titinopathies that might be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Martin Rees
- Randall Centre for Cell and Molecular Biophysics, Muscle Biophysics, King's College London BHF Centre of Research Excellence, London, UK
| | - Roksana Nikoopour
- Randall Centre for Cell and Molecular Biophysics, Muscle Biophysics, King's College London BHF Centre of Research Excellence, London, UK
| | - Atsushi Fukuzawa
- Randall Centre for Cell and Molecular Biophysics, Muscle Biophysics, King's College London BHF Centre of Research Excellence, London, UK
| | - Ay Lin Kho
- Randall Centre for Cell and Molecular Biophysics, Muscle Biophysics, King's College London BHF Centre of Research Excellence, London, UK
| | - Miguel A Fernandez-Garcia
- Department of Paediatric Neurology, Evelina Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - Elizabeth Wraige
- Department of Paediatric Neurology, Evelina Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - Istvan Bodi
- Department of Clinical Neuropathology, King's College Hospital, London, UK
| | | | - Özkan Özdemir
- Centre for Molecular Medicine, University of Cologne, Cologne, Germany
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Hülya-Sevcan Daimagüler
- Centre for Molecular Medicine, University of Cologne, Cologne, Germany
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Mark Pfuhl
- Randall Centre for Cell and Molecular Biophysics, Muscle Biophysics, King's College London BHF Centre of Research Excellence, London, UK
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre of Research Excellence, London, UK
| | - Mark Holt
- Randall Centre for Cell and Molecular Biophysics, Muscle Biophysics, King's College London BHF Centre of Research Excellence, London, UK
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre of Research Excellence, London, UK
| | - Birgit Brandmeier
- Randall Centre for Cell and Molecular Biophysics, Muscle Biophysics, King's College London BHF Centre of Research Excellence, London, UK
| | - Sarah Grover
- Randall Centre for Cell and Molecular Biophysics, Muscle Biophysics, King's College London BHF Centre of Research Excellence, London, UK
| | - Joël Fluss
- Pediatric Neurology Unit, Paediatrics Subspecialties Service, Geneva Children's Hospital, Geneva, Switzerland
| | - Cheryl Longman
- West of Scotland Regional Genetics Service, Laboratory Medicine Building, Queen Elizabeth University Hospital, Glasgow, UK
| | | | - Emma Matthews
- MRC Neuromuscular Centre, National Hospital for Neurology and Neurosurgery, Queen's Square, London, UK
| | - Michael Hanna
- MRC Neuromuscular Centre, National Hospital for Neurology and Neurosurgery, Queen's Square, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, Great Ormond Street Hospital Trust, London, UK
| | - Anna Sarkozy
- Dubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children, London, UK
| | - Rahul Phadke
- Dubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children, London, UK
| | - Ros Quinlivan
- Dubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children, London, UK
| | - Emily C Oates
- Dubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children, London, UK
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sidney, Australia
- Kids Neuroscience Centre, Kids Research, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Thiel
- Department of Genetics, University of Erlangen, Erlangen, Germany
| | - Jens Reimann
- Muscle Laboratory, Department of Neurology, University of Bonn Medical Centre, Bonn, Germany
| | - Nicol Voermans
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Corrie Erasmus
- Department of Paediatric Neurology, Radboud University, Nijmegen, The Netherlands
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Chaminda Konersman
- UCSD, Rady Children's Hospital, and VA San Diego Healthcare System, San Diego, USA
| | | | - Shane McKee
- Northern Ireland Regional Genetics Service, Belfast City Hospital, Belfast, UK
| | - Sandya Tirupathi
- Department of Paediatric Neurology, Royal Belfast Hospital for Sick Children, Belfast, UK
| | - Steven A Moore
- Department of Pathology, The University of Iowa, Iowa City, IA, USA
| | | | - Elke Hobbiebrunken
- Department of Paediatric Neurology, University of Göttingen, Göttingen, Germany
| | | | - Isabelle Richard
- Genethon and UMR_S951, INSERM, Université Evry, Université Paris Saclay, Evry, 91002, Evry, France
| | - Peter Van den Bergh
- Neuromuscular Reference Centre, Department of Neurology, University Hospital Saint-Luc, Brussels, Belgium
| | | | - Sebahattin Cirak
- Centre for Molecular Medicine, University of Cologne, Cologne, Germany
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Centre for Rare Diseases (ZSEK), University of Cologne, Cologne, Germany
| | - Ana Ferreiro
- Basic and Translational Myology Laboratory, Université de Paris, Paris, France
- Centre de Référence Des Maladies Neuromusculaires, APHP, Institut of Myology, GHU Pitié Salpêtrière- Charles Foix, Paris, France
| | - Heinz Jungbluth
- Randall Centre for Cell and Molecular Biophysics, Muscle Biophysics, King's College London BHF Centre of Research Excellence, London, UK
- Department of Paediatric Neurology, Evelina Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK
- Department of Clinical and Basic Neuroscience, IoPPN, King's College London, London, UK
| | - Mathias Gautel
- Randall Centre for Cell and Molecular Biophysics, Muscle Biophysics, King's College London BHF Centre of Research Excellence, London, UK.
| |
Collapse
|
28
|
Jiang H, Hooper C, Kelly M, Steeples V, Simon JN, Beglov J, Azad AJ, Leinhos L, Bennett P, Ehler E, Kalisch-Smith JI, Sparrow DB, Fischer R, Heilig R, Isackson H, Ehsan M, Patone G, Huebner N, Davies B, Watkins H, Gehmlich K. Functional analysis of a gene-edited mouse model to gain insights into the disease mechanisms of a titin missense variant. Basic Res Cardiol 2021; 116:14. [PMID: 33637999 PMCID: PMC7910237 DOI: 10.1007/s00395-021-00853-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/10/2021] [Indexed: 11/03/2022]
Abstract
Titin truncating variants are a well-established cause of cardiomyopathy; however, the role of titin missense variants is less well understood. Here we describe the generation of a mouse model to investigate the underlying disease mechanism of a previously reported titin A178D missense variant identified in a family with non-compaction and dilated cardiomyopathy. Heterozygous and homozygous mice carrying the titin A178D missense variant were characterised in vivo by echocardiography. Heterozygous mice had no detectable phenotype at any time point investigated (up to 1 year). By contrast, homozygous mice developed dilated cardiomyopathy from 3 months. Chronic adrenergic stimulation aggravated the phenotype. Targeted transcript profiling revealed induction of the foetal gene programme and hypertrophic signalling pathways in homozygous mice, and these were confirmed at the protein level. Unsupervised proteomics identified downregulation of telethonin and four-and-a-half LIM domain 2, as well as the upregulation of heat shock proteins and myeloid leukaemia factor 1. Loss of telethonin from the cardiac Z-disc was accompanied by proteasomal degradation; however, unfolded telethonin accumulated in the cytoplasm, leading to a proteo-toxic response in the mice.We show that the titin A178D missense variant is pathogenic in homozygous mice, resulting in cardiomyopathy. We also provide evidence of the disease mechanism: because the titin A178D variant abolishes binding of telethonin, this leads to its abnormal cytoplasmic accumulation. Subsequent degradation of telethonin by the proteasome results in proteasomal overload, and activation of a proteo-toxic response. The latter appears to be a driving factor for the cardiomyopathy observed in the mouse model.
Collapse
Affiliation(s)
- He Jiang
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Charlotte Hooper
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Matthew Kelly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Violetta Steeples
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Jillian N Simon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Julia Beglov
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Amar J Azad
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Lisa Leinhos
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Pauline Bennett
- Randall Centre for Cell and Molecular Biophysics, School of Cardiovascular Medicine and Sciences, King's College London BHF Centre of Research Excellence, London, UK
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics, School of Cardiovascular Medicine and Sciences, King's College London BHF Centre of Research Excellence, London, UK
| | | | - Duncan B Sparrow
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Roman Fischer
- Nuffield Department of Clinical Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
| | - Raphael Heilig
- Nuffield Department of Clinical Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
| | - Henrik Isackson
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
- Department of Medical Cell Biology, Integrative Physiology, Uppsala University, Uppsala, Sweden
| | - Mehroz Ehsan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Giannino Patone
- Max Delbrueck Centre for Molecular Medicine, Berlin, Germany
| | - Norbert Huebner
- Max Delbrueck Centre for Molecular Medicine, Berlin, Germany
| | - Benjamin Davies
- Transgenic Core, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK.
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
29
|
Santiago CF, Huttner IG, Fatkin D. Mechanisms of TTNtv-Related Dilated Cardiomyopathy: Insights from Zebrafish Models. J Cardiovasc Dev Dis 2021; 8:jcdd8020010. [PMID: 33504111 PMCID: PMC7912658 DOI: 10.3390/jcdd8020010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a common heart muscle disorder characterized by ventricular dilation and contractile dysfunction that is associated with significant morbidity and mortality. New insights into disease mechanisms and strategies for treatment and prevention are urgently needed. Truncating variants in the TTN gene, which encodes the giant sarcomeric protein titin (TTNtv), are the most common genetic cause of DCM, but exactly how TTNtv promote cardiomyocyte dysfunction is not known. Although rodent models have been widely used to investigate titin biology, they have had limited utility for TTNtv-related DCM. In recent years, zebrafish (Danio rerio) have emerged as a powerful alternative model system for studying titin function in the healthy and diseased heart. Optically transparent embryonic zebrafish models have demonstrated key roles of titin in sarcomere assembly and cardiac development. The increasing availability of sophisticated imaging tools for assessment of heart function in adult zebrafish has revolutionized the field and opened new opportunities for modelling human genetic disorders. Genetically modified zebrafish that carry a human A-band TTNtv have now been generated and shown to spontaneously develop DCM with age. This zebrafish model will be a valuable resource for elucidating the phenotype modifying effects of genetic and environmental factors, and for exploring new drug therapies.
Collapse
Affiliation(s)
- Celine F. Santiago
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (C.F.S.); (I.G.H.)
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Inken G. Huttner
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (C.F.S.); (I.G.H.)
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (C.F.S.); (I.G.H.)
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- Cardiology Department, St. Vincent’s Hospital, Darlinghurst, NSW 2010, Australia
- Correspondence:
| |
Collapse
|
30
|
Huang C, Hou C, Ijaz M, Yan T, Li X, Li Y, Zhang D. Proteomics discovery of protein biomarkers linked to meat quality traits in post-mortem muscles: Current trends and future prospects: A review. Trends Food Sci Technol 2020. [DOI: 10.1016/j.tifs.2020.09.030] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
31
|
Blondelle J, Biju A, Lange S. The Role of Cullin-RING Ligases in Striated Muscle Development, Function, and Disease. Int J Mol Sci 2020; 21:E7936. [PMID: 33114658 PMCID: PMC7672578 DOI: 10.3390/ijms21217936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
The well-orchestrated turnover of proteins in cross-striated muscles is one of the fundamental processes required for muscle cell function and survival. Dysfunction of the intricate protein degradation machinery is often associated with development of cardiac and skeletal muscle myopathies. Most muscle proteins are degraded by the ubiquitin-proteasome system (UPS). The UPS involves a number of enzymes, including E3-ligases, which tightly control which protein substrates are marked for degradation by the proteasome. Recent data reveal that E3-ligases of the cullin family play more diverse and crucial roles in cross striated muscles than previously anticipated. This review highlights some of the findings on the multifaceted functions of cullin-RING E3-ligases, their substrate adapters, muscle protein substrates, and regulatory proteins, such as the Cop9 signalosome, for the development of cross striated muscles, and their roles in the etiology of myopathies.
Collapse
Affiliation(s)
- Jordan Blondelle
- Department of Medicine, University of California, La Jolla, CA 92093, USA
| | - Andrea Biju
- Department of Medicine, University of California, La Jolla, CA 92093, USA
| | - Stephan Lange
- Department of Medicine, University of California, La Jolla, CA 92093, USA
- Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| |
Collapse
|
32
|
Swist S, Unger A, Li Y, Vöge A, von Frieling-Salewsky M, Skärlén Å, Cacciani N, Braun T, Larsson L, Linke WA. Maintenance of sarcomeric integrity in adult muscle cells crucially depends on Z-disc anchored titin. Nat Commun 2020; 11:4479. [PMID: 32900999 PMCID: PMC7478974 DOI: 10.1038/s41467-020-18131-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 08/04/2020] [Indexed: 12/14/2022] Open
Abstract
The giant protein titin is thought to be required for sarcomeric integrity in mature myocytes, but direct evidence for this hypothesis is limited. Here, we describe a mouse model in which Z-disc-anchored TTN is depleted in adult skeletal muscles. Inactivation of TTN causes sarcomere disassembly and Z-disc deformations, force impairment, myocyte de-stiffening, upregulation of TTN-binding mechanosensitive proteins and activation of protein quality-control pathways, concomitant with preferential loss of thick-filament proteins. Interestingly, expression of the myosin-bound Cronos-isoform of TTN, generated from an alternative promoter not affected by the targeting strategy, does not prevent deterioration of sarcomere formation and maintenance. Finally, we demonstrate that loss of Z-disc-anchored TTN recapitulates muscle remodeling in critical illness ‘myosinopathy’ patients, characterized by TTN-depletion and loss of thick filaments. We conclude that full-length TTN is required to integrate Z-disc and A-band proteins into the mature sarcomere, a function that is lost when TTN expression is pathologically lowered. Titin is considered an integrator of muscle cell proteins but direct evidence is limited. Here, titin is inactivated in adult mouse muscles, which causes sarcomere disassembly, protein mis-expression and force impairment, recapitulating key alterations in critical illness myopathy patient muscles.
Collapse
Affiliation(s)
- Sandra Swist
- Department of Systems Physiology, Ruhr University Bochum, D-44780, Bochum, Germany.
| | - Andreas Unger
- Institute of Physiology II, University of Munster, D-48149, Munster, Germany
| | - Yong Li
- Institute of Physiology II, University of Munster, D-48149, Munster, Germany
| | - Anja Vöge
- Department of Systems Physiology, Ruhr University Bochum, D-44780, Bochum, Germany
| | | | - Åsa Skärlén
- Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institute, SE-171 77, Stockholm, Sweden
| | - Nicola Cacciani
- Department of Physiology and Pharmacology, Karolinska Institute, SE-171 77, Stockholm, Sweden
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, D-61231, Bad Nauheim, Germany
| | - Lars Larsson
- Department of Physiology and Pharmacology, Karolinska Institute, SE-171 77, Stockholm, Sweden
| | - Wolfgang A Linke
- Institute of Physiology II, University of Munster, D-48149, Munster, Germany.
| |
Collapse
|
33
|
Lange S, Pinotsis N, Agarkova I, Ehler E. The M-band: The underestimated part of the sarcomere. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118440. [PMID: 30738787 PMCID: PMC7023976 DOI: 10.1016/j.bbamcr.2019.02.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/16/2019] [Accepted: 02/05/2019] [Indexed: 12/20/2022]
Abstract
The sarcomere is the basic unit of the myofibrils, which mediate skeletal and cardiac Muscle contraction. Two transverse structures, the Z-disc and the M-band, anchor the thin (actin and associated proteins) and thick (myosin and associated proteins) filaments to the elastic filament system composed of titin. A plethora of proteins are known to be integral or associated proteins of the Z-disc and its structural and signalling role in muscle is better understood, while the molecular constituents of the M-band and its function are less well defined. Evidence discussed here suggests that the M-band is important for managing force imbalances during active muscle contraction. Its molecular composition is fine-tuned, especially as far as the structural linkers encoded by members of the myomesin family are concerned and depends on the specific mechanical characteristics of each particular muscle fibre type. Muscle activity signals from the M-band to the nucleus and affects transcription of sarcomeric genes, especially via serum response factor (SRF). Due to its important role as shock absorber in contracting muscle, the M-band is also more and more recognised as a contributor to muscle disease.
Collapse
Affiliation(s)
- Stephan Lange
- Biomedical Research Facility 2, School of Medicine, University of California, San Diego, Medical Sciences Research Bldg, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA; University of Gothenburg, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden
| | - Nikos Pinotsis
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, Malet Street, London WC1E 7HX, UK
| | - Irina Agarkova
- InSphero, Wagistrasse 27, CH-8952 Schlieren, Switzerland
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK; School of Cardiovascular Medicine and Sciences, British Heart Foundation Research Excellence Centre, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK.
| |
Collapse
|
34
|
Radke MH, Polack C, Methawasin M, Fink C, Granzier HL, Gotthardt M. Deleting Full Length Titin Versus the Titin M-Band Region Leads to Differential Mechanosignaling and Cardiac Phenotypes. Circulation 2020; 139:1813-1827. [PMID: 30700140 DOI: 10.1161/circulationaha.118.037588] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Titin is a giant elastic protein that spans the half-sarcomere from Z-disk to M-band. It acts as a molecular spring and mechanosensor and has been linked to striated muscle disease. The pathways that govern titin-dependent cardiac growth and contribute to disease are diverse and difficult to dissect. METHODS To study titin deficiency versus dysfunction, the authors generated and compared striated muscle specific knockouts (KOs) with progressive postnatal loss of the complete titin protein by removing exon 2 (E2-KO) or an M-band truncation that eliminates proper sarcomeric integration, but retains all other functional domains (M-band exon 1/2 [M1/2]-KO). The authors evaluated cardiac function, cardiomyocyte mechanics, and the molecular basis of the phenotype. RESULTS Skeletal muscle atrophy with reduced strength, severe sarcomere disassembly, and lethality from 2 weeks of age were shared between the models. Cardiac phenotypes differed considerably: loss of titin leads to dilated cardiomyopathy with combined systolic and diastolic dysfunction-the absence of M-band titin to cardiac atrophy and preserved function. The elastic properties of M1/2-KO cardiomyocytes are maintained, while passive stiffness is reduced in the E2-KO. In both KOs, we find an increased stress response and increased expression of proteins linked to titin-based mechanotransduction (CryAB, ANKRD1, muscle LIM protein, FHLs, p42, Camk2d, p62, and Nbr1). Among them, FHL2 and the M-band signaling proteins p62 and Nbr1 are exclusively upregulated in the E2-KO, suggesting a role in the differential pathology of titin truncation versus deficiency of the full-length protein. The differential stress response is consistent with truncated titin contributing to the mechanical properties in M1/2-KOs, while low titin levels in E2-KOs lead to reduced titin-based stiffness and increased strain on the remaining titin molecules. CONCLUSIONS Progressive depletion of titin leads to sarcomere disassembly and atrophy in striated muscle. In the complete knockout, remaining titin molecules experience increased strain, resulting in mechanically induced trophic signaling and eventually dilated cardiomyopathy. The truncated titin in M1/2-KO helps maintain the passive properties and thus reduces mechanically induced signaling. Together, these findings contribute to the molecular understanding of why titin mutations differentially affect cardiac growth and have implications for genotype-phenotype relations that support a personalized medicine approach to the diverse titinopathies.
Collapse
Affiliation(s)
- Michael H Radke
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany (M.H.R., C.P., C.F., M.G.).,DZHK: German Centre for Cardiovascular Research, Partner Site, Berlin, Germany (M.H.R., M.G.)
| | - Christopher Polack
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany (M.H.R., C.P., C.F., M.G.)
| | - Mei Methawasin
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (M.M., H.G.). The current affiliation for P.S. and T.S. is Department of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Germany
| | - Claudia Fink
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany (M.H.R., C.P., C.F., M.G.)
| | - Henk L Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (M.M., H.G.). The current affiliation for P.S. and T.S. is Department of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Germany
| | - Michael Gotthardt
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany (M.H.R., C.P., C.F., M.G.).,DZHK: German Centre for Cardiovascular Research, Partner Site, Berlin, Germany (M.H.R., M.G.)
| |
Collapse
|
35
|
Chatziefthimiou SD, Hornburg P, Sauer F, Mueller S, Ugurlar D, Xu ER, Wilmanns M. Structural diversity in the atomic resolution 3D fingerprint of the titin M-band segment. PLoS One 2019; 14:e0226693. [PMID: 31856237 PMCID: PMC6922384 DOI: 10.1371/journal.pone.0226693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 12/02/2019] [Indexed: 11/18/2022] Open
Abstract
In striated muscles, molecular filaments are largely composed of long protein chains with extensive arrays of identically folded domains, referred to as “beads-on-a-string”. It remains a largely unresolved question how these domains have developed a unique molecular profile such that each carries out a distinct function without false-positive readout. This study focuses on the M-band segment of the sarcomeric protein titin, which comprises ten identically folded immunoglobulin domains. Comparative analysis of high-resolution structures of six of these domains ‒ M1, M3, M4, M5, M7, and M10 ‒ reveals considerable structural diversity within three distinct loops and a non-conserved pattern of exposed cysteines. Our data allow to structurally interpreting distinct pathological readouts that result from titinopathy-associated variants. Our findings support general principles that could be used to identify individual structural/functional profiles of hundreds of identically folded protein domains within the sarcomere and other densely crowded cellular environments.
Collapse
Affiliation(s)
| | - Philipp Hornburg
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Florian Sauer
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Simone Mueller
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Deniz Ugurlar
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Emma-Ruoqi Xu
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Matthias Wilmanns
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
- University Hamburg Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
36
|
Blondelle J, Marrocco V, Clark M, Desmond P, Myers S, Nguyen J, Wright M, Bremner S, Pierantozzi E, Ward S, Estève E, Sorrentino V, Ghassemian M, Lange S. Murine obscurin and Obsl1 have functionally redundant roles in sarcolemmal integrity, sarcoplasmic reticulum organization, and muscle metabolism. Commun Biol 2019; 2:178. [PMID: 31098411 PMCID: PMC6509138 DOI: 10.1038/s42003-019-0405-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/28/2019] [Indexed: 12/19/2022] Open
Abstract
Biological roles of obscurin and its close homolog Obsl1 (obscurin-like 1) have been enigmatic. While obscurin is highly expressed in striated muscles, Obsl1 is found ubiquitously. Accordingly, obscurin mutations have been linked to myopathies, whereas mutations in Obsl1 result in 3M-growth syndrome. To further study unique and redundant functions of these closely related proteins, we generated and characterized Obsl1 knockouts. Global Obsl1 knockouts are embryonically lethal. In contrast, skeletal muscle-specific Obsl1 knockouts show a benign phenotype similar to obscurin knockouts. Only deletion of both proteins and removal of their functional redundancy revealed their roles for sarcolemmal stability and sarcoplasmic reticulum organization. To gain unbiased insights into changes to the muscle proteome, we analyzed tibialis anterior and soleus muscles by mass spectrometry, uncovering additional changes to the muscle metabolism. Our analyses suggest that all obscurin protein family members play functions for muscle membrane systems.
Collapse
Affiliation(s)
- Jordan Blondelle
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Valeria Marrocco
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Madison Clark
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Patrick Desmond
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Stephanie Myers
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Jim Nguyen
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Matthew Wright
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Shannon Bremner
- Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Enrico Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, 53100 Italy
| | - Samuel Ward
- Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Eric Estève
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
- Université Grenoble Alpes, HP2, Grenoble, 38706 France
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, 53100 Italy
| | - Majid Ghassemian
- Department of Chemistry and Biochemistry, University of California, San Diego, 92093 CA USA
| | - 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
| |
Collapse
|
37
|
Koser F, Loescher C, Linke WA. Posttranslational modifications of titin from cardiac muscle: how, where, and what for? FEBS J 2019; 286:2240-2260. [PMID: 30989819 PMCID: PMC6850032 DOI: 10.1111/febs.14854] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/27/2019] [Accepted: 04/13/2019] [Indexed: 12/11/2022]
Abstract
Titin is a giant elastic protein expressed in the contractile units of striated muscle cells, including the sarcomeres of cardiomyocytes. The last decade has seen enormous progress in our understanding of how titin molecular elasticity is modulated in a dynamic manner to help cardiac sarcomeres adjust to the varying hemodynamic demands on the heart. Crucial events mediating the rapid modulation of cardiac titin stiffness are post‐translational modifications (PTMs) of titin. In this review, we first recollect what is known from earlier and recent work on the molecular mechanisms of titin extensibility and force generation. The main goal then is to provide a comprehensive overview of current insight into the relationship between titin PTMs and cardiomyocyte stiffness, notably the effect of oxidation and phosphorylation of titin spring segments on titin stiffness. A synopsis is given of which type of oxidative titin modification can cause which effect on titin stiffness. A large part of the review then covers the mechanically relevant phosphorylation sites in titin, their location along the elastic segment, and the protein kinases and phosphatases known to target these sites. We also include a detailed coverage of the complex changes in phosphorylation at specific titin residues, which have been reported in both animal models of heart disease and in human heart failure, and their correlation with titin‐based stiffness alterations. Knowledge of the relationship between titin PTMs and titin elasticity can be exploited in the search for therapeutic approaches aimed at softening the pathologically stiffened myocardium in heart failure patients.
Collapse
|
38
|
Picard B, Gagaoua M, Al Jammas M, Bonnet M. Beef tenderness and intramuscular fat proteomic biomarkers: Effect of gender and rearing practices. J Proteomics 2019; 200:1-10. [PMID: 30894324 DOI: 10.1016/j.jprot.2019.03.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 12/24/2022]
Abstract
This study analyzed the effect of gender on the abundances of 20 protein biomarkers of tenderness and/or intramuscular fat content in five muscles: Longissimus thoracis, previously identified as biomarkers of tenderness and/or intramuscular Semimembranosus, Rectus abdominis, Triceps brachii and Semitendinosus, from cows and steers of the Protected Designation Origin Maine Anjou. The protein abundances were quantified using Reverse Phase Protein Array with specific validated antibodies. Among the 20 studied proteins, the abundance of 8 biomarkers involved in energetic metabolism, contraction and cellular stress, was different according to gender. The gender effect was different depending on the muscle type with greater abundances in Semitendinosus, Rectus abdominis and Longissimus thoracis muscles. On the basis of animal characteristics and rearing factors, three rearing practices classes were identified for cows. Among the factors, fattening duration modified the abundance of 12 proteins mainly in Triceps brachii muscle. A positive correlation between the abundance of the small HSP20 and slaughter age was observed in the 5 muscles. Two proteins, Four and a half LIM domains 1 (FHL1) and Glycogen phosphorylase (PYGB) appeared to be muscle, gender and rearing practices independent. These results constitute valuable data to understand how to manage beef quality by controlling these different factors. SIGNIFICANCE: This study is the first to compare the relative abundance of 20 proteins previously identified as biomarkers of tenderness and/or intramuscular fat (IMF) content of beef meat between cows and steers among 5 different muscles. Its originality is in the use of Reverse Phase Protein Array for fast quantification of the proteins and the integration of data from rearing factors, carcass characteristics and biomarkers of meat qualities. The findings provide evidence for modulating biomarker levels by controlling the choice of animal type and rearing factors according to the type of muscle that would produce animals with the desired meat qualities.
Collapse
Affiliation(s)
- Brigitte Picard
- Université Clermont Auvergne, INRA, VetAgro Sup, UMR Herbivores, Saint-Genès-Champanelle, France.
| | - Mohammed Gagaoua
- Université Clermont Auvergne, INRA, VetAgro Sup, UMR Herbivores, Saint-Genès-Champanelle, France.
| | - Marwa Al Jammas
- Université Clermont Auvergne, INRA, VetAgro Sup, UMR Herbivores, Saint-Genès-Champanelle, France
| | - Muriel Bonnet
- Université Clermont Auvergne, INRA, VetAgro Sup, UMR Herbivores, Saint-Genès-Champanelle, France
| |
Collapse
|
39
|
Shufelt CL, Pacheco C, Tweet MS, Miller VM. Sex-Specific Physiology and Cardiovascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1065:433-454. [PMID: 30051400 PMCID: PMC6768431 DOI: 10.1007/978-3-319-77932-4_27] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Sex differences in cardiovascular diseases can be classified as those which are specific to one sex and those that differ in incidence, prevalence, etiology, symptomatology, response to treatment, morbidity, and mortality in one sex compared to the other. All sex differences in cardiovascular conditions have their basis in the combined expression of genetic and hormonal differences between women and men. This chapter addresses how understanding basic mechanisms of hormone responses, imaging diagnostics, and integration of genomics and proteomics has advanced diagnosis and improved outcomes for cardiovascular conditions, apart from those related to pregnancy that are more prevalent in women. These conditions include obstructive coronary artery disease, coronary microvascular dysfunction, spontaneous coronary artery dissection, diseases of the cardiac muscle including heart failure and takotsubo cardiomyopathy, and conditions related to neurovascular dysregulation including hot flashes and night sweats associated with menopause and effects of exogenous hormones on vascular function. Improvement in technologies allowing for noninvasive assessment of neuronally mediated vascular reactivity will further improve our understanding of the basic etiology of the neurovascular disorders. Consideration of sex, hormonal status, and pregnancy history in diagnosis and treatment protocols will improve prevention and outcomes of cardiovascular disease in women as they age.
Collapse
Affiliation(s)
- Chrisandra L Shufelt
- Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Smidt Heart Insititute, Los Angeles, CA, USA.
| | - Christine Pacheco
- Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Smidt Heart Insititute, Los Angeles, CA, USA
| | - Marysia S Tweet
- Department of Cardiovascular Medicine, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Virginia M Miller
- Surgery and Physiology, Women's Health Research Center, College of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
40
|
Liu Z, Han S, Wang Y, Cui C, Zhu Q, Jiang X, Yang C, Du H, Yu C, Li Q, He H, Shen X, Chen Y, Zhang Y, Ye L, Zhang Z, Li D, Zhao X, Yin H. The LIM-Only Protein FHL2 is involved in Autophagy to Regulate the Development of Skeletal Muscle Cell. Int J Biol Sci 2019; 15:838-846. [PMID: 30906214 PMCID: PMC6429013 DOI: 10.7150/ijbs.31371] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/21/2019] [Indexed: 12/15/2022] Open
Abstract
Scope: Four and a half LIM domain protein 2 (FHL2) is a LIM domain protein expressed in muscle tissue whose deletion is causative of myopathies. Although FHL2 has a confirmed important role in muscle development, its autophagy-related function in muscle differentiation has not been fully determined. Methods: C2C12 cells were treated with FHL2-konwdown or FHL2-overexpression. The morphology of C2C12 cells was observed by transmission electron microscopy. The mRNA and protein abundances of muscle related genes and autophagy related genes were measured by RT-PCR and western blot. Immunofluorescence and co-immunoprecipitation assay were used to verify the interaction between FHL2 and LC3 protein. Results: FHL2 silencing reduced LC3-Ⅱ protein expression and the amount of LC3 that co-immunoprecipitated with FHL2, indicating that FHL2 interacts with LC3-Ⅱ in the formation of autophagosomes. Moreover, the expression of muscle development marker genes such as MyoD1 and MyoG was lower in FHL2-silenced C2C12 cells but not in FHL2-overexpressing C2C12 cells. Electron microscopy analysis revealed large empty autophagosomes in FHL2-silenced myoblasts, while flow cytometry suggested that FHL2 silencing made cells more vulnerable to staurosporine-induced cell death. Conclusion: These results suggest that FHL2 interacts with LC3-Ⅱ in autophagosome formation to regulate the development of muscle cells.
Collapse
Affiliation(s)
- Zihao Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Shunshun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Can Cui
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Qing Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Xiaosong Jiang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, 610066, PR China
| | - Chaowu Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, 610066, PR China
| | - Huarui Du
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, 610066, PR China
| | - Chunlin Yu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, 610066, PR China
| | - Qingyun Li
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, 610066, PR China
| | - Haorong He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Xiaoxu Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Yuqi Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Yao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Lin Ye
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Zhichao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Diyan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Xiaoling Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Huadong Yin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| |
Collapse
|
41
|
Lynch JM, Dolman AJ, Guo C, Dolan K, Xiang C, Reda S, Li B, Prasanna G. Mutant myocilin impacts sarcomere ultrastructure in mouse gastrocnemius muscle. PLoS One 2018; 13:e0206801. [PMID: 30395621 PMCID: PMC6218065 DOI: 10.1371/journal.pone.0206801] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 10/19/2018] [Indexed: 11/19/2022] Open
Abstract
Myocilin (MYOC) is the gene with mutations most common in glaucoma. In the eye, MYOC is in trabecular meshwork, ciliary body, and retina. Other tissues with high MYOC transcript levels are skeletal muscle and heart. To date, the function of wild-type MYOC remains unknown and how mutant MYOC causes high intraocular pressure and glaucoma is ambiguous. By investigating mutant MYOC in a non-ocular tissue we hoped to obtain novel insight into mutant MYOC pathology. For this study, we utilized a transgenic mouse expressing human mutant MYOC Y437H protein and we examined its skeletal (gastrocnemius) muscle phenotype. Electron micrographs showed that sarcomeres in the skeletal muscle of mutant CMV-MYOC-Y437H mice had multiple M-bands. Western blots of soluble muscle lysates from transgenics indicated a decrease in two M-band proteins, myomesin 1 (MYOM1) and muscle creatine kinase (CKM). Immunoprecipitation identified CKM as a MYOC binding partner. Our results suggest that binding of mutant MYOC to CKM is changing sarcomere ultrastructure and this may adversely impact muscle function. We speculate that a person carrying the mutant MYOC mutation will likely have a glaucoma phenotype and may also have undiagnosed muscle ailments or vice versa, both of which will have to be monitored and treated.
Collapse
MESH Headings
- Animals
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Disease Models, Animal
- Eye Proteins/genetics
- Eye Proteins/metabolism
- Female
- Gene Expression
- Glaucoma, Open-Angle/genetics
- Glaucoma, Open-Angle/metabolism
- Glaucoma, Open-Angle/pathology
- Glycoproteins/genetics
- Glycoproteins/metabolism
- Humans
- Intraocular Pressure/genetics
- Male
- Mice
- Mice, Mutant Strains
- Mice, Transgenic
- Microscopy, Electron, Transmission
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/ultrastructure
- Mutant Proteins/genetics
- Mutant Proteins/metabolism
- Mutation
- Myocardium/metabolism
- Phenotype
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Sarcomeres/genetics
- Sarcomeres/metabolism
- Sarcomeres/ultrastructure
- Trabecular Meshwork/metabolism
- Trabecular Meshwork/ultrastructure
Collapse
Affiliation(s)
- Jeffrey M. Lynch
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
- * E-mail:
| | - Andrew J. Dolman
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Chenying Guo
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Katie Dolan
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Chuanxi Xiang
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Samir Reda
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Bing Li
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Ganesh Prasanna
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| |
Collapse
|
42
|
Gagaoua M, Bonnet M, De Koning L, Picard B. Reverse Phase Protein array for the quantification and validation of protein biomarkers of beef qualities: The case of meat color from Charolais breed. Meat Sci 2018; 145:308-319. [DOI: 10.1016/j.meatsci.2018.06.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 06/26/2018] [Accepted: 06/26/2018] [Indexed: 12/25/2022]
|
43
|
Four and a half LIM domain protein signaling and cardiomyopathy. Biophys Rev 2018; 10:1073-1085. [PMID: 29926425 DOI: 10.1007/s12551-018-0434-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 06/06/2018] [Indexed: 01/10/2023] Open
Abstract
Four and a half LIM domain (FHL) protein family members, FHL1 and FHL2, are multifunctional proteins that are enriched in cardiac muscle. Although they both localize within the cardiomyocyte sarcomere (titin N2B), they have been shown to have important yet unique functions within the context of cardiac hypertrophy and disease. Studies in FHL1-deficient mice have primarily uncovered mitogen-activated protein kinase (MAPK) scaffolding functions for FHL1 as part of a novel biomechanical stretch sensor within the cardiomyocyte sarcomere, which acts as a positive regulator of pressure overload-mediated cardiac hypertrophy. New data have highlighted a novel role for the serine/threonine protein phosphatase (PP5) as a deactivator of the FHL1-based biomechanical stretch sensor, which has implications in not only cardiac hypertrophy but also heart failure. In contrast, studies in FHL2-deficient mice have primarily uncovered an opposing role for FHL2 as a negative regulator of adrenergic-mediated signaling and cardiac hypertrophy, further suggesting unique functions targeted by FHL proteins in the "stressed" cardiomyocyte. In this review, we provide current knowledge of the role of FHL1 and FHL2 in cardiac muscle as it relates to their actions in cardiac hypertrophy and cardiomyopathy. A specific focus will be to dissect the pathways and protein-protein interactions that underlie FHLs' signaling role in cardiac hypertrophy as well as provide a comprehensive list of FHL mutations linked to cardiac disease, using evidence gained from genetic mouse models and human genetic studies.
Collapse
|
44
|
Wang C, Chu J, Fu L, Wang Y, Zhao F, Zhou D. iTRAQ-based quantitative proteomics reveals the biochemical mechanism of cold stress adaption of razor clam during controlled freezing-point storage. Food Chem 2018; 247:73-80. [DOI: 10.1016/j.foodchem.2017.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/21/2017] [Accepted: 12/04/2017] [Indexed: 12/23/2022]
|
45
|
Savarese M, Sarparanta J, Vihola A, Udd B, Hackman P. Increasing Role of Titin Mutations in Neuromuscular Disorders. J Neuromuscul Dis 2018; 3:293-308. [PMID: 27854229 PMCID: PMC5123623 DOI: 10.3233/jnd-160158] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The TTN gene with 363 coding exons encodes titin, a giant muscle protein spanning from the Z-disk to the M-band within the sarcomere. Mutations in the TTN gene have been associated with different genetic disorders, including hypertrophic and dilated cardiomyopathy and several skeletal muscle diseases. Before the introduction of next generation sequencing (NGS) methods, the molecular analysis of TTN has been laborious, expensive and not widely used, resulting in a limited number of mutations identified. Recent studies however, based on the use of NGS strategies, give evidence of an increasing number of rare and unique TTN variants. The interpretation of these rare variants of uncertain significance (VOUS) represents a challenge for clinicians and researchers. The main aim of this review is to describe the wide spectrum of muscle diseases caused by TTN mutations so far determined, summarizing the molecular findings as well as the clinical data, and to highlight the importance of joint efforts to respond to the challenges arising from the use of NGS. An international collaboration through a clinical and research consortium and the development of a single accessible database listing variants in the TTN gene, identified by high throughput approaches, may be the key to a better assessment of titinopathies and to systematic genotype– phenotype correlation studies.
Collapse
Affiliation(s)
- Marco Savarese
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Jaakko Sarparanta
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland.,Albert Einstein College of Medicine, Departments of Medicine- Endocrinology and Molecular Pharmacology, Bronx, NY, USA
| | - Anna Vihola
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Bjarne Udd
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland.,Neuromuscular Research Center, University of Tampere and Tampere University Hospital, Tampere, Finland.,Department of Neurology, Vaasa Central Hospital, Vaasa, Finland
| | - Peter Hackman
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
| |
Collapse
|
46
|
Wang L, Geist J, Grogan A, Hu LYR, Kontrogianni-Konstantopoulos A. Thick Filament Protein Network, Functions, and Disease Association. Compr Physiol 2018; 8:631-709. [PMID: 29687901 PMCID: PMC6404781 DOI: 10.1002/cphy.c170023] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sarcomeres consist of highly ordered arrays of thick myosin and thin actin filaments along with accessory proteins. Thick filaments occupy the center of sarcomeres where they partially overlap with thin filaments. The sliding of thick filaments past thin filaments is a highly regulated process that occurs in an ATP-dependent manner driving muscle contraction. In addition to myosin that makes up the backbone of the thick filament, four other proteins which are intimately bound to the thick filament, myosin binding protein-C, titin, myomesin, and obscurin play important structural and regulatory roles. Consistent with this, mutations in the respective genes have been associated with idiopathic and congenital forms of skeletal and cardiac myopathies. In this review, we aim to summarize our current knowledge on the molecular structure, subcellular localization, interacting partners, function, modulation via posttranslational modifications, and disease involvement of these five major proteins that comprise the thick filament of striated muscle cells. © 2018 American Physiological Society. Compr Physiol 8:631-709, 2018.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | | |
Collapse
|
47
|
Guo W, Sun M. RBM20, a potential target for treatment of cardiomyopathy via titin isoform switching. Biophys Rev 2018; 10:15-25. [PMID: 28577155 PMCID: PMC5803173 DOI: 10.1007/s12551-017-0267-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/16/2017] [Indexed: 12/18/2022] Open
Abstract
Cardiomyopathy, also known as heart muscle disease, is an unfavorable condition leading to alterations in myocardial contraction and/or impaired ability of ventricular filling. The onset and development of cardiomyopathy have not currently been well defined. Titin is a giant multifunctional sarcomeric filament protein that provides passive stiffness to cardiomyocytes and has been implicated to play an important role in the origin and development of cardiomyopathy and heart failure. Titin-based passive stiffness can be mainly adjusted by isoform switching and post-translational modifications in the spring regions. Recently, genetic mutations of TTN have been identified that can also contribute to variable passive stiffness, though the detailed mechanisms remain unclear. In this review, we will discuss titin isoform switching as it relates to alternative splicing during development stages and differences between species and muscle types. We provide an update on the regulatory mechanisms of TTN splicing controlled by RBM20 and cover the roles of TTN splicing in adjusting the diastolic stiffness and systolic compliance of the healthy and the failing heart. Finally, this review attempts to provide future directions for RBM20 as a potential target for pharmacological intervention in cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Wei Guo
- Animal Science, University of Wyoming, Laramie, WY, 82071, USA.
- Center for Cardiovascular Research and Integrative Medicine, University of Wyoming, Laramie, WY, 82071, USA.
| | - Mingming Sun
- Animal Science, University of Wyoming, Laramie, WY, 82071, USA
- Center for Cardiovascular Research and Integrative Medicine, University of Wyoming, Laramie, WY, 82071, USA
| |
Collapse
|
48
|
Protein phosphatase 5 regulates titin phosphorylation and function at a sarcomere-associated mechanosensor complex in cardiomyocytes. Nat Commun 2018; 9:262. [PMID: 29343782 PMCID: PMC5772059 DOI: 10.1038/s41467-017-02483-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 12/04/2017] [Indexed: 12/14/2022] Open
Abstract
Serine/threonine protein phosphatase 5 (PP5) is ubiquitously expressed in eukaryotic cells; however, its function in cardiomyocytes is unknown. Under basal conditions, PP5 is autoinhibited, but enzymatic activity rises upon binding of specific factors, such as the chaperone Hsp90. Here we show that PP5 binds and dephosphorylates the elastic N2B-unique sequence (N2Bus) of titin in cardiomyocytes. Using various binding and phosphorylation tests, cell-culture manipulation, and transgenic mouse hearts, we demonstrate that PP5 associates with N2Bus in vitro and in sarcomeres and is antagonistic to several protein kinases, which phosphorylate N2Bus and lower titin-based passive tension. PP5 is pathologically elevated and likely contributes to hypo-phosphorylation of N2Bus in failing human hearts. Furthermore, Hsp90-activated PP5 interacts with components of a sarcomeric, N2Bus-associated, mechanosensor complex, and blocks mitogen-activated protein-kinase signaling in this complex. Our work establishes PP5 as a compartmentalized, well-controlled phosphatase in cardiomyocytes, which regulates titin properties and kinase signaling at the myofilaments. Protein phosphatase 5 (PP5) is expressed in many cell types but its role in cardiomyocytes is unknown. Here the authors show that PP5 binds and dephosphorylates elastic titin in cardiac sarcomeres, and that PP5 is increased in heart failure, reducing cardiomyocyte compliance.
Collapse
|
49
|
Pang SM, Le S, Yan J. Mechanical responses of the mechanosensitive unstructured domains in cardiac titin. Biol Cell 2017; 110:65-76. [DOI: 10.1111/boc.201700061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 11/26/2022]
Affiliation(s)
- Si Ming Pang
- Mechanobiology Institute; National University of Singapore; 117411 Singapore
| | - Shimin Le
- Mechanobiology Institute; National University of Singapore; 117411 Singapore
- Department of Physics; National University of Singapore; 117542 Singapore
| | - Jie Yan
- Mechanobiology Institute; National University of Singapore; 117411 Singapore
- Department of Physics; National University of Singapore; 117542 Singapore
- Centre for Bioimaging Sciences; National University of Singapore; 117546 Singapore
| |
Collapse
|
50
|
Ehsan M, Jiang H, L Thomson K, Gehmlich K. When signalling goes wrong: pathogenic variants in structural and signalling proteins causing cardiomyopathies. J Muscle Res Cell Motil 2017; 38:303-316. [PMID: 29119312 PMCID: PMC5742121 DOI: 10.1007/s10974-017-9487-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/28/2017] [Indexed: 12/20/2022]
Abstract
Cardiomyopathies are a diverse group of cardiac disorders with distinct phenotypes, depending on the proteins and pathways affected. A substantial proportion of cardiomyopathies are inherited and those will be the focus of this review article. With the wide application of high-throughput sequencing in the practice of clinical genetics, the roles of novel genes in cardiomyopathies are recognised. Here, we focus on a subgroup of cardiomyopathy genes [TTN, FHL1, CSRP3, FLNC and PLN, coding for Titin, Four and a Half LIM domain 1, Muscle LIM Protein, Filamin C and Phospholamban, respectively], which, despite their diverse biological functions, all have important signalling functions in the heart, suggesting that disturbances in signalling networks can contribute to cardiomyopathies.
Collapse
Affiliation(s)
- Mehroz Ehsan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - He Jiang
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Kate L Thomson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK.
| |
Collapse
|