1
|
Yakupova EI, Abramicheva PA, Rogachevsky VV, Shishkova EA, Bocharnikov AD, Plotnikov EY, Vikhlyantsev IM. Cardiac titin isoforms: Practice in interpreting results of electrophoretic analysis. Methods 2025; 236:17-25. [PMID: 39993454 DOI: 10.1016/j.ymeth.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025] Open
Abstract
The sarcomeric giant protein titin affects the passive elasticity of the heart muscle and is crucial for proper cardiac function, including diastolic relaxation of the left ventricle. A useful common method for studying titin is electrophoretic analysis which can be used to examine the distribution of its isoforms in the heart. There are 5 titin parameters that can be analyzed: the N2BA/N2B isoforms ratio, the T2/T1 bands ratio, Cronos isoform content, NT isoform content, the total titin-to-myosin heavy chain (TT/MHC) ratio. These parameters can only be assessed through electrophoresis of giant proteins. It is known that these parameters are related to various biomolecular processes in muscle cells, such as providing of elastic properties, turnover, contraction, and maintaining a highly ordered sarcomere structure. In this review, we discuss the diagnostic potential of electrophoretic visualization of cardiac titin changes in various human heart diseases and animal models of physiological adaptations or pathologies.
Collapse
Affiliation(s)
- Elmira I Yakupova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Polina A Abramicheva
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Vadim V Rogachevsky
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino 142290 Moscow Region, Russia
| | - Elena A Shishkova
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino 142290 Moscow Region, Russia
| | - Alexey D Bocharnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Egor Y Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Ivan M Vikhlyantsev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia; Pushchino Branch of the Federal State Budgetary Educational Institution of Higher Education «Russian Biotechnological University (BIOTECH University)», Russia.
| |
Collapse
|
2
|
Coppens S, Deconinck N, Sullivan P, Smolnikov A, Clayton JS, Griffin KR, Jones KJ, Vilain CN, Kadhim H, Bryen SJ, Faiz F, Waddell LB, Evesson FJ, Bakshi M, Pinner JR, Charlton A, Brammah S, Graf NS, Krivanek M, Tay CG, Foulds NC, Illingworth MA, Thomas NH, Ellard S, Mazanti I, Park S, French CE, Brewster J, Belteki G, Hoodbhoy S, Allinson K, Krishnakumar D, Baynam G, Wood BM, Ward M, Vijayakumar K, Syed A, Murugan A, Majumdar A, Scurr IJ, Splitt MP, Moldovan C, de Silva DC, Senanayake K, Gardeitchik T, Arens Y, Cooper ST, Laing NG, Raymond FL, Jungbluth H, Kamsteeg E, Manzur A, Corley SM, Ravenscroft G, Wilkins MR, Cowley MJ, Pinese M, Phadke R, Davis MR, Muntoni F, Oates EC. Congenital Titinopathy: Comprehensive Characterization of the Most Severe End of the Disease Spectrum. Ann Neurol 2025; 97:611-628. [PMID: 39853809 PMCID: PMC11889535 DOI: 10.1002/ana.27087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 01/26/2025]
Abstract
Congenital titinopathy has recently emerged as one of the most common congenital muscle disorders. OBJECTIVE To better understand the presentation and clinical needs of the under-characterized extreme end of the congenital titinopathy severity spectrum. METHODS We comprehensively analyzed the clinical, imaging, pathology, autopsy, and genetic findings in 15 severely affected individuals from 11 families. RESULTS Prenatal features included hypokinesia or akinesia and growth restriction. Six pregnancies were terminated. Nine infants were born at or near term with severe-to-profound weakness and required resuscitation. Seven died following withdrawal of life support. Two surviving children require ongoing respiratory support. Most cohort members had at least 1 disease-causing variant predicted to result in some near-normal-length titin expression. The exceptions, from 2 unrelated families, had homozygous truncating variants predicted to induce complete nonsense mediated decay. However, subsequent analyses suggested that the causative variant in each family had an additional previously unrecognized impact on splicing likely to result in some near-normal-length titin expression. This impact was confirmed by minigene assay for 1 variant. INTERPRETATION This study confirms the clinical variability of congenital titinopathy. Severely affected individuals succumb prenatally/during infancy, whereas others survive into adulthood. It is likely that this variability is because of differences in the amount and/or length of expressed titin. If confirmed, analysis of titin expression could facilitate clinical prediction and increasing expression might be an effective treatment strategy. Our findings also further-support the hypothesis that some near-normal-length titin expression is essential to early prenatal survival. Sometimes expression of normal/near-normal-length titin is due to disease-causing variants having an additional impact on splicing. ANN NEUROL 2025;97:611-628.
Collapse
Affiliation(s)
- Sandra Coppens
- Hopital Erasme, ULB Center of Human GeneticsUniversité Libre de BruxellesBrusselsBelgium
| | - Nicolas Deconinck
- Department of Paediatric Neurology, Neuromuscular Reference CenterHôpital Universitaire des Enfants Reine Fabiola, Université Libre de BruxellesBrusselsBelgium
| | - Patricia Sullivan
- Children's Cancer Institute, Lowy Cancer CentreUniversity of New South WalesSydneyNew South WalesAustralia
| | - Andrei Smolnikov
- School of Biotechnology and Biomolecular SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Joshua S. Clayton
- Harry Perkins Institute of Medical Research, QEII Medical CentreNedlandsWestern AustraliaAustralia
- Centre for Medical ResearchThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Kaitlyn R. Griffin
- School of Biotechnology and Biomolecular SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Kristi J. Jones
- Kids Neuroscience Centre, Kids ResearchThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Faculty of Medicine and HealthThe University of SydneyWestmeadNew South WalesAustralia
| | - Catheline N. Vilain
- Hopital Erasme, ULB Center of Human GeneticsUniversité Libre de BruxellesBrusselsBelgium
| | - Hazim Kadhim
- Neuropathology Unit (Anatomic Pathology Service) and Reference Center for Neuromuscular PathologyCHU Brugmann‐HUDERF, Université Libre de BruxellesBrusselsBelgium
| | - Samantha J. Bryen
- Kids Neuroscience Centre, Kids ResearchThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Faculty of Medicine and HealthThe University of SydneyWestmeadNew South WalesAustralia
| | - Fathimath Faiz
- Department of Diagnostic Genomics, PathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Leigh B. Waddell
- Kids Neuroscience Centre, Kids ResearchThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Faculty of Medicine and HealthThe University of SydneyWestmeadNew South WalesAustralia
| | - Frances J. Evesson
- Kids Neuroscience Centre, Kids ResearchThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Faculty of Medicine and HealthThe University of SydneyWestmeadNew South WalesAustralia
- Functional NeuromicsChildren's Medical Research InstituteWestmeadNew South WalesAustralia
| | - Madhura Bakshi
- Department of Clinical GeneticsLiverpool HospitalLiverpoolNew South WalesAustralia
| | - Jason R. Pinner
- Department of Medical GenomicsRoyal Prince Alfred Hospital, The University of SydneyCamperdownNew South WalesAustralia
| | - Amanda Charlton
- Department of HistopathologyThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
| | - Susan Brammah
- Electron Microscope Unit, Department of Anatomical PathologyConcord Repatriation General HospitalConcordNew South WalesAustralia
| | - Nicole S. Graf
- Department of HistopathologyThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
| | - Michael Krivanek
- Department of HistopathologyThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
| | - Chee Geap Tay
- Division of Paediatric Neurology, Department of Paediatrics, Faculty of MedicineUniversity of MalayaKuala LumpurMalaysia
| | - Nicola C. Foulds
- Wessex Clinical Genetics ServiceUniversity Hospital Southampton NHS Foundation TrustSouthamptonUK
| | - Marjorie A. Illingworth
- Department of Paediatric NeurologyUniversity Hospital Southampton NHS Foundation TrustSouthamptonUK
| | - Neil H. Thomas
- Department of Paediatric NeurologyUniversity Hospital Southampton NHS Foundation TrustSouthamptonUK
| | - Sian Ellard
- College of Medicine and Health, University of Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation TrustExeterUK
| | - Ingrid Mazanti
- Department of Cellular PathologyUniversity Hospital Southampton NHS Foundation TrustSouthamptonUK
| | - Soo‐Mi Park
- Department of Clinical GeneticsCambridge University Hospital NHS Foundation TrustCambridgeUK
| | - Courtney E. French
- Department of Medical GeneticsCambridge Institute for Medical Research, University of CambridgeCambridgeUK
| | - Jennifer Brewster
- Department of Fetomaternal MedicineCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Gusztav Belteki
- Neonatal Intensive Care UnitCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Shazia Hoodbhoy
- Neonatal Intensive Care UnitCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Kieren Allinson
- Department of HistopathologyCambridge University Hospitals Foundation TrustCambridgeUK
| | - Deepa Krishnakumar
- Department of Paediatric NeurologyCambridge University Hospitals Foundation TrustCambridgeUK
| | - Gareth Baynam
- Genetic Services of Western AustraliaKing Edward Memorial HospitalPerthWestern AustraliaAustralia
| | | | - Michelle Ward
- Genetic Services of Western AustraliaKing Edward Memorial HospitalPerthWestern AustraliaAustralia
| | - Kayal Vijayakumar
- Department of Paediatric NeurologyUniversity Hospitals Bristol NHS Foundation TrustBristolUK
| | - Amber Syed
- Department of Paediatric NeurologyUniversity Hospitals Bristol NHS Foundation TrustBristolUK
| | - Archana Murugan
- Department of Paediatric NeurologyUniversity Hospitals Bristol NHS Foundation TrustBristolUK
| | - Anirban Majumdar
- Department of Paediatric NeurologyUniversity Hospitals Bristol NHS Foundation TrustBristolUK
| | - Ingrid J. Scurr
- Department of Clinical GeneticsUniversity Hospitals Bristol NHS Foundation TrustBristolUK
| | - Miranda P. Splitt
- Northern Genetics ServiceInstitute of Genetic MedicineNewcastle upon TyneUK
| | - Corina Moldovan
- Department of PathologyNewcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Deepthi C. de Silva
- Department of Physiology, Faculty of MedicineUniversity of KelaniyaRagamaSri Lanka
| | - Kumudu Senanayake
- Department of HistopathologyCastle Street Hospital for WomenColomboSri Lanka
| | - Thatjana Gardeitchik
- Department of Human GeneticsRadboud University Medical CenterNijmegenthe Netherlands
| | - Yvonne Arens
- Department of Clinical GeneticsMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Sandra T. Cooper
- Kids Neuroscience Centre, Kids ResearchThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Faculty of Medicine and HealthThe University of SydneyWestmeadNew South WalesAustralia
- Functional NeuromicsChildren's Medical Research InstituteWestmeadNew South WalesAustralia
| | - Nigel G. Laing
- Harry Perkins Institute of Medical Research, QEII Medical CentreNedlandsWestern AustraliaAustralia
- Centre for Medical ResearchThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - F. Lucy Raymond
- Department of Medical GeneticsCambridge Institute for Medical Research, University of CambridgeCambridgeUK
| | - Heinz Jungbluth
- Department of Paediatric Neurology, Neuromuscular ServiceEvelina's Children Hospital, Guy's and St. Thomas' Hospital NHS Foundation TrustLondonUK
- Randall Division for Cell and Molecular Biophysics, Muscle Signalling SectionKing's College LondonLondonUK
- Department of Basic and Clinical NeuroscienceIoPPN, King's College LondonLondonUK
| | - Erik‐Jan Kamsteeg
- Department of Human GeneticsRadboud University Medical CenterNijmegenthe Netherlands
| | - Adnan Manzur
- Great Ormond Street Hospital for ChildrenNHS Foundation Trust, Dubowitz Neuromuscular CentreLondonUK
| | - Susan M. Corley
- Systems Biology Initiative, School of Biotechnology and Biomolecular ScienceUniversity of New South WalesSydneyNew South WalesAustralia
| | - Gianina Ravenscroft
- Harry Perkins Institute of Medical Research, QEII Medical CentreNedlandsWestern AustraliaAustralia
- Centre for Medical ResearchThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Marc R. Wilkins
- School of Biotechnology and Biomolecular SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Mark J. Cowley
- Children's Cancer Institute, Lowy Cancer CentreUniversity of New South WalesSydneyNew South WalesAustralia
| | - Mark Pinese
- Children's Cancer Institute, Lowy Cancer CentreUniversity of New South WalesSydneyNew South WalesAustralia
| | - Rahul Phadke
- Great Ormond Street Hospital for ChildrenNHS Foundation Trust, Dubowitz Neuromuscular CentreLondonUK
- Division of NeuropathologyUCL Institute of Neurology, The National Hospital for Neurology and NeurosurgeryLondonUK
| | - Mark R. Davis
- Department of Diagnostic Genomics, PathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Francesco Muntoni
- Great Ormond Street Hospital for ChildrenNHS Foundation Trust, Dubowitz Neuromuscular CentreLondonUK
| | - Emily C. Oates
- School of Biotechnology and Biomolecular SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Department of NeurologySydney Children's HospitalSydneyNew South WalesAustralia
| |
Collapse
|
3
|
Wang Y, Ye J, Liu X, Zhang Z, Shang F, Qi X, Zhang Y, Du J, Sun H, Xu J, Chen H, Yu M, Le S. Mechanically weak and highly dynamic state of mechanosensitive titin Ig domains induced by proline isomerization. Nat Commun 2025; 16:2771. [PMID: 40113761 PMCID: PMC11926396 DOI: 10.1038/s41467-025-57989-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
Titin, essential for mechano-homeostasis in cardiac and skeletal sarcomere, contains numerous mechanosensitive immunoglobulin-like (Ig) domains in its I-band region. However, how proline isomerization and cysteine-mediated disulfide bond collectively regulate Ig domain dynamics within the physiological force range remains unclear. Here, we use single-molecule force spectroscopy to quantify the proximal Ig1 domain, revealing that proline isomerization leads to two native states-trans and cis states-with distinct mechanical and thermal stabilities. The trans-Ig1 unfolds at forces of ~ 5 pN, which is over 50 pN lower than that of cis-Ig1, and unfolds 1000 times faster under physiological forces. Furthermore, such proline induced dual-state is likely shared feature across majority of I-band Ig domains. Additionally, reduced cis- and trans-Ig1 exhibit catch-slip bond unfolding, while oxidized forms display slip-catch-slip unfolding. This study offers insight into effective modulation of proline isomerization and disulfide bond in regulating mechanosensitive proteins within the physiological force range.
Collapse
Affiliation(s)
- Yukai Wang
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, Xiamen, China
- Department of Biochemistry and Division of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqing Ye
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, Xiamen, China
| | - Xian Liu
- Department of Biochemistry and Division of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhuwei Zhang
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, Xiamen, China
| | - Fei Shang
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, Xiamen, China
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xingyu Qi
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, Xiamen, China
| | - Yuhang Zhang
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, Xiamen, China
| | - Jingyi Du
- Department of Biochemistry and Division of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Sun
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, Xiamen, China
- Center of Biomedical Physics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Jiashu Xu
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, Xiamen, China
| | - Hu Chen
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, Xiamen, China.
- Center of Biomedical Physics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
| | - Miao Yu
- Department of Biochemistry and Division of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Shimin Le
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, Xiamen, China.
| |
Collapse
|
4
|
Zhu C, Bishop T, Gregorich ZR, Guo W. Titin is a new factor regulating arterial stiffness through vascular smooth muscle cell tone in male rats. Physiol Rep 2025; 13:e70270. [PMID: 40119572 PMCID: PMC11928681 DOI: 10.14814/phy2.70270] [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: 11/12/2024] [Revised: 02/03/2025] [Accepted: 02/28/2025] [Indexed: 03/24/2025] Open
Abstract
Arterial stiffness is a robust predictor of cardiovascular disease and mortality. As such, there is substantial interest in uncovering its causal factors for the development of targeted treatments to regulate arterial stiffness. The elastic protein titin is a key determinant of myocardial stiffness, yet whether it plays a role in regulating arterial stiffness is unknown. In this study, we aimed to investigate the role of titin in vascular smooth muscle cell (VSMC) and overall arterial stiffness. To do this, we took advantage of rats lacking RNA binding motif 20 (RBM20), the primary splicing regulator of titin, in striated muscles. Using this model, we demonstrate that RBM20 regulates titin isoform expression in smooth muscle, with loss of the protein leading to the expression of larger titin isoforms. We show that the expression of larger titin reduces the stiffness of VSMCs. While decreased titin-based VSMC stiffness did not affect baseline arterial stiffness, we found that arterial stiffness was reduced in response to a challenge with the potent vasoconstrictor angiotensin II (Ang II). The observed reduction in arterial stiffness following Ang II treatment was not the result of changes in either the extracellular matrix or myofilaments. We further show that the expression of a larger titin isoform ameliorates cardiac remodeling caused by Ang II-associated hypertension. In summary, our study provides the first evidence that titin regulates VSMC stiffness, which is relevant for arterial stiffness in the context of elevated blood pressure. Furthermore, our data provide proof-of-concept evidence that targeting RBM20 to reduce arterial stiffness through titin isoform switching may benefit aging- or hypertension-associated arterial stiffness and vascular diseases.
Collapse
Affiliation(s)
- Chaoqun Zhu
- Department of Animal Sciences, University of Wyoming, Laramie, Wyoming, USA
| | | | - Zachery R Gregorich
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Cardiovascular Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Wei Guo
- Department of Animal Sciences, University of Wyoming, Laramie, Wyoming, USA
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Cardiovascular Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
5
|
van der Pijl R, Nusayr E, Strom J, Slater R, Gohlke J, Hourani Z, Saripalli C, Kolb J, Hermanson K, Brynnel O, Smith JE, Labeit S, Methawasin M, Granzier H. Importance of N2BA Titin in Maintaining Cardiac Homeostasis and Its Role in Dilated Cardiomyopathy. Circ Heart Fail 2025; 18:e012083. [PMID: 39932400 PMCID: PMC11905908 DOI: 10.1161/circheartfailure.124.012083] [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: 06/05/2024] [Accepted: 12/20/2024] [Indexed: 03/15/2025]
Abstract
BACKGROUND TTN (titin) is the third myofilament type of the cardiac sarcomere and performs important functions that include generating passive tension. Changes in TTN expression are associated with cardiac dysfunction, and TTN is one of the main genes linked to dilated cardiomyopathy (DCM). DCM is frequently associated with changes in the expression of N2BA (compliant cardiac TTN isoform), 1 of the 2 major TTN isoforms found in the heart (the other isoform being the N2B [stiff cardiac TTN isoform]). Whether altered expression of N2BA TTN causes DCM or is a secondary change remains unclear. METHODS Here, we present a mouse model, the TtnΔ112-158 model, which specifically shortens the proline, glutamate, valine, lysine region of the N2BA isoform. RESULTS Echocardiography and pressure-volume analysis revealed a DCM phenotype characterized by systolic dysfunction and dilation. RNA sequencing studies showed the absence of proline, glutamate, valine, lysine exons, as expected, but also reduced expressions of exons specific to the N2BA isoform of TTN. Protein studies revealed a reduction in the overall expression level of the N2BA isoform with a concomitant increase in N2B TTN, with preserved TT (total TTN) levels. Passive tension was modestly increased in the TtnΔ112-158 model. Western blotting revealed that the N2BA TTN-associated protein MARP1 (muscle ankyrin repeat protein 1) is downregulated during both the pre-DCM and DCM phase. Downregulation of MARP1 coincided with the downregulation of the transcription factor Gata-4 (GATA binding protein 4), an MARP1-regulating and interacting protein, which is associated with DCM development. CONCLUSIONS Thus, N2BA TTN is essential for maintaining cardiac health, and perturbed N2BA-MARP1 signaling contributes to DCM development.
Collapse
Affiliation(s)
- Robbert van der Pijl
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - Eyad Nusayr
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - Joshua Strom
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - Rebecca Slater
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - Jochen Gohlke
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - Zaynab Hourani
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - Chandra Saripalli
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - Justin Kolb
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - Kyra Hermanson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - Odhin Brynnel
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - John E. Smith
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - Siegfried Labeit
- Medical Faculty Mannheim, Department of Integrative Pathophysiology, DZHK Partner Site Mannheim-Heidelberg, Germany (S.L.)
| | - Mei Methawasin
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson (R.v.d.P., E.N., J.S., R.S., J.G., Z.H., C.S., J.K., K.H., O.B., J.E.S., M.M., H.G.)
| |
Collapse
|
6
|
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
|
7
|
Hüttemeister J, Rudolph F, Radke MH, Fink C, Friedrich D, Preibisch S, Falcke M, Wagner E, Lehnart SE, Gotthardt M. Visualizing sarcomere and cellular dynamics in skeletal muscle to improve cell therapies. eLife 2024; 13:e95597. [PMID: 39688479 DOI: 10.7554/elife.95597] [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: 12/27/2023] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The giant striated muscle protein titin integrates into the developing sarcomere to form a stable myofilament system that is extended as myocytes fuse. The logistics underlying myofilament assembly and disassembly have started to emerge with the possibility to follow labeled sarcomere components. Here, we generated the mCherry knock-in at titin's Z-disk to study skeletal muscle development and remodeling. We find titin's integration into the sarcomere tightly regulated and its unexpected mobility facilitating a homogeneous distribution of titin after cell fusion - an integral part of syncytium formation and maturation of skeletal muscle. In adult mCherry-titin mice, treatment of muscle injury by implantation of titin-eGFP myoblasts reveals how myocytes integrate, fuse, and contribute to the continuous myofilament system across cell boundaries. Unlike in immature primary cells, titin proteins are retained at the proximal nucleus and do not diffuse across the whole syncytium with implications for future cell-based therapies of skeletal muscle disease.
Collapse
Affiliation(s)
- Judith Hüttemeister
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| | - Franziska Rudolph
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Michael H Radke
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Claudia Fink
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Dhana Friedrich
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Stephan Preibisch
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Martin Falcke
- Computational Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Eva Wagner
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
| | - Stephan E Lehnart
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Gotthardt
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
8
|
Briganti F, Wang Z. Alternative Splicing in the Heart: The Therapeutic Potential of Regulating the Regulators. Int J Mol Sci 2024; 25:13023. [PMID: 39684734 PMCID: PMC11641712 DOI: 10.3390/ijms252313023] [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: 10/29/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Alternative splicing allows a single gene to produce a variety of protein isoforms. Changes in splicing isoform usage characterize virtually every stage of the differentiation process and define the physiological differences between cardiomyocytes with different function, at different stages of development, and pathological function. Recent identification of cardiac splicing factors provided insights into the mechanisms underlying alternative splicing and revealed how these splicing factors impact functional properties of the heart. Alterations of the splicing of sarcomeric genes, cell signaling proteins, and ion channels have been associated with the development of pathological conditions such as cardiomyopathy and arrhythmia. RBM20, RBM24, PTBP1, RBFOX, and QKI play key roles in cardiac development and pathology. A better understanding of their regulation will yield insights into healthy cardiac development and inform the development of molecular therapeutics.
Collapse
Affiliation(s)
- Francesca Briganti
- Division of Genetics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
- Division of Cardiology, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Zilu Wang
- Division of Genetics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
9
|
Abel N, Schupp T, Schmitt A, Reinhardt M, Lau F, Weidner K, Ayoub M, Mashayekhi K, Akin I, Behnes M. Left ventricular diastolic dysfunction in patients with heart failure with mildly reduced ejection fraction. Int J Cardiol 2024; 414:132386. [PMID: 39079587 DOI: 10.1016/j.ijcard.2024.132386] [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: 03/01/2024] [Revised: 06/27/2024] [Accepted: 07/17/2024] [Indexed: 08/06/2024]
Abstract
OBJECTIVE This study investigates the prevalence and prognostic impact of diastolic dysfunction (DD) in patients hospitalized with heart failure (HF) with mildly reduced ejection fraction (HFmrEF) in sinus rhythm. BACKGROUND Data regarding the prognostic impact of DD in patients with HFmrEF is limited. METHODS From 2016 to 2022, all patients hospitalized with HFmrEF (i.e., left ventricular ejection fraction 41-49% and signs and/or symptoms of HF) were retrospectively included at one institution. Patients with DD were compared to patients without (i.e., non-DD), further risk stratification was performed according to the severity of DD. The primary endpoint was all-cause mortality at 30 months (interquartile range (IQR) 15-61 months), key secondary endpoint was rehospitalization for worsening HF. RESULTS From a total of 1154 patients (median age 68 years, 68% males) hospitalized with HFmrEF, concomitant DD was present in 72% (grade I: 56%, grade II: 14%, grade III: 2%). Patients with DD were older (71 years vs. 65 years; p = 0.001) and presented with higher rates of cardiovascular comorbidities. The presence of DD was not associated with the risk of long-term all-cause mortality (adjusted HR = 0.815; 95% CI 0.612-1.085; p = 0.161) or HF-related rehospitalization (adjusted HR = 0.736; 95% CI 0.442-1.225; p = 0.238). Furthermore, the outcome did not differ in patients with more advanced stages of DD. CONCLUSION DD is commonly prevalent in patients with HFmrEF, but not associated with long-term prognosis.
Collapse
Affiliation(s)
- Noah Abel
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Tobias Schupp
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Alexander Schmitt
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Marielen Reinhardt
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Felix Lau
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Kathrin Weidner
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Mohamed Ayoub
- Division of Cardiology and Angiology, Heart Center University of Bochum, Bad Oeynhausen 32545, Germany
| | - Kambis Mashayekhi
- Department of Internal Medicine and Cardiology, MediClin Heart Centre Lahr, Lahr 77933, Germany
| | - Ibrahim Akin
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Michael Behnes
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| |
Collapse
|
10
|
Furukawa N, Matsui H, Sunaga H, Nagata K, Hirayama M, Obinata H, Yokoyama T, Ohno K, Kurabayashi M, Koitabashi N. Sacubitril/valsartan improves diastolic left ventricular stiffness with increased titin phosphorylation via cGMP-PKG activation in diabetic mice. Sci Rep 2024; 14:25081. [PMID: 39443532 PMCID: PMC11499646 DOI: 10.1038/s41598-024-75757-8] [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: 03/31/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Titin, a giant sarcomeric protein, regulates diastolic left ventricular (LV) passive stiffness as a molecular spring and could be a therapeutic target for diastolic dysfunction. Sacubitril/valsartan (Sac/Val), an angiotensin receptor neprilysin inhibitor, has been shown to benefit patients with heart failure with preserved ejection fraction. The effect of Sac/Val is thought to be due to the enhancement of the cGMP/PKG pathway via natriuretic peptide. In this study, the effects of Sac/Val on LV diastolic dysfunction are demonstrated in a mouse diabetic cardiomyopathy model focusing on titin phosphorylation. Sac/Val-treated diabetic mice showed a greater increase in myocardial levels of cGMP-PKG than Val-treated and control mice. Conductance catheter analysis showed a significant reduction in LV stiffness in diabetic mice, but not in non-diabetic mice. Notably, diastolic LV stiffness was significantly reduced in Sac/Val-treated diabetic hearts compared with Val-treated or vehicle-treated diabetic mice. The phosphorylation level of titin (N2B), which determines passive stiffness and modulates active contraction, was higher in Sac/Val-treated hearts compared with Val-treated hearts in diabetic mice. Given that alteration of titin phosphorylation through PKG contributes to myocardial stiffness, the beneficial effects of Sac/Val in heart failure might be partly attributed to the induction of titin phosphorylation.
Collapse
Affiliation(s)
- Nozomi Furukawa
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22, Showa-Machi, Maebashi, Gunma, 371-8511, Japan
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Matsui
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Japan
| | - Hiroaki Sunaga
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22, Showa-Machi, Maebashi, Gunma, 371-8511, Japan
- Center for Liberal Arts and Sciences, Ashikaga University, Ashikaga, Japan
| | - Kohzo Nagata
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaaki Hirayama
- Department of Occupational Therapy, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Hideru Obinata
- Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tomoyuki Yokoyama
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Japan
| | - Kinji Ohno
- Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, Nisshin, Japan
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiko Kurabayashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22, Showa-Machi, Maebashi, Gunma, 371-8511, Japan
| | - Norimichi Koitabashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22, Showa-Machi, Maebashi, Gunma, 371-8511, Japan.
| |
Collapse
|
11
|
Shaftoe JB, Geddes-McAlister J, Gillis TE. Integrated cellular response of the zebrafish (Danio rerio) heart to temperature change. J Exp Biol 2024; 227:jeb247522. [PMID: 39091230 DOI: 10.1242/jeb.247522] [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: 02/21/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
A decrease in environmental temperature represents a challenge to the cardiovascular system of ectotherms. To gain insight into the cellular changes that occur during cold exposure and cold acclimation we characterized the cardiac phosphoproteome and proteome of zebrafish following 24 h or 1 week exposure to 20°C from 27°C; or at multiple points during 6 weeks of acclimation to 20°C from 27°C. Our results indicate that cold exposure causes an increase in mitogen-activated protein kinase signalling, the activation of stretch-sensitive pathways, cellular remodelling via ubiquitin-dependent pathways and changes to the phosphorylation state of proteins that regulate myofilament structure and function including desmin and troponin T. Cold acclimation (2-6 weeks) led to a decrease in multiple components of the electron transport chain through time, but an increase in proteins for lipid transport, lipid metabolism, the incorporation of polyunsaturated fatty acids into membranes and protein turnover. For example, there was an increase in the levels of apolipoprotein C, prostaglandin reductase-3 and surfeit locus protein 4, involved in lipid transport, lipid metabolism and lipid membrane remodelling. Gill opercular movements suggest that oxygen utilization during cold acclimation is reduced. Neither the amount of food consumed relative to body mass nor body condition was affected by acclimation. These results suggest that while oxygen uptake was reduced, energy homeostasis was maintained. This study highlights that the response of zebrafish to a decrease in temperature is dynamic through time and that investment in the proteomic response increases with the duration of exposure.
Collapse
Affiliation(s)
- Jared B Shaftoe
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - Jennifer Geddes-McAlister
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - Todd E Gillis
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| |
Collapse
|
12
|
Zhazykbayeva S, Budde H, Kaçmaz M, Zemedie Y, Osman H, Hassoun R, Jaquet K, Akin I, El-Battrawy I, Herwig M, Hamdani N. Exploring PKG signaling as a therapeutic avenue for pressure overload, ischemia, and HFpEF. Expert Opin Ther Targets 2024; 28:857-873. [PMID: 39329430 DOI: 10.1080/14728222.2024.2400093] [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: 01/10/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
INTRODUCTION Heart failure (HF) is a complex and heterogeneous syndrome resulting from any diastolic or systolic dysfunction of the cardiac muscle. In addition to comorbid conditions, pressure overload, and myocardial ischemia are associated with cardiac remodeling which manifests as extracellular matrix (ECM) perturbations, impaired cellular responses, and subsequent ventricular dysfunction. AREAS COVERED The current review discusses the main aspects of the cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) pathway (cGMP-PKG) pathway modulators and highlights the promising outcomes of its novel pharmacological boosters. EXPERT OPINION Among several signaling pathways involved in the pathogenesis of pressure overload, ischemia and HF with preserved ejection fraction (HFpEF) is cGMP-PKG pathway. This pathway plays a pivotal role in the regulation of cardiac contractility, and modulation of cGMP-PKG signaling, contributing to the development of the diseases. Ventricular cardiomyocytes of HF patients and animal models are known to exhibit reduced cGMP levels and disturbed cGMP signaling including hypophosphorylation of PKG downstream targets. However, restoration of cGMP-PKG signaling improves cardiomyocyte function and promotes cardioprotective effects.
Collapse
Affiliation(s)
- S Zhazykbayeva
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - H Budde
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - M Kaçmaz
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Intézet címe Semmelweis University, Budapest, Hungary
| | - Y Zemedie
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - H Osman
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - R Hassoun
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - K Jaquet
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - I Akin
- Medical University Mannheim, Medical Faculty, Mannheim University, Heidelberg, Germany
| | - I El-Battrawy
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University, Bochum, Germany
| | - M Herwig
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - N Hamdani
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Intézet címe Semmelweis University, Budapest, Hungary
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University, Bochum, Germany
- Department of Physiology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
13
|
Lakomkin VL, Abramov AA, Prosvirnin AV, Tereshchenko AS, Arutunan GK, Samko AN, Kapelko VI. The Structure of Left Ventricular Relaxation in Case of Ventriculography. KARDIOLOGIIA 2024; 64:32-38. [PMID: 39262351 DOI: 10.18087/cardio.2024.8.n2640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/11/2024] [Indexed: 09/13/2024]
Abstract
AIM To study the relaxation structure of the left ventricle (LV) in patients who underwent ventriculography. MATERIAL AND METHODS LV ventriculography was performed in 37 patients. Before catheterization, echocardiography was performed in each patient. In 6 patients, the LV ejection fraction (EF) was below 40%; these patients with systolic dysfunction were not included in the study. In 31 patients, the LV EF was higher than 50%. In this group, 13 patients had NYHA functional class (FC) 2-3 chronic heart failure (CHF); the rest of the patients had FC 1 CHF. Eighteen of 31 patients had stable ischemic heart disease; 50% of these patients had a history of myocardial infarction; the rest of the patients had hypertension and atrial and ventricular arrhythmias. The dynamics of the LV pressure decrease was analyzed from the moment of the maximum rate of pressure drop, which usually coincides with the closure of the aortic valves. The pressure drop curve was logarithmized with natural logarithms and divided into 4-5 sections with different degrees of curve slope. The relaxation time constant was calculated for each section. Its inverse value characterizes the relaxation time constant (tau). RESULTS In 31 patients with LV EF 52-60%, three types of the dynamics of the relaxation rate constant were identified during the pressure decrease in the isovolumic phase: in 9 patients, the isovolumic relaxation constant (IRC) steadily increased as the pressure decreased; in 13 patients, it continuously decreased; and in 9 patients, the dynamics of IRC change was intermediate, with an initial increase followed by a decrease. CONCLUSION In diastolic dysfunction, one group of patients had an adaptation type associated with an increase in the LV wall elasticity, while the other group had a different type of adaptation associated with its decrease. Each type has advantages and disadvantages. This is probably due to changes in the structure of the sarcomeric protein connectin (titin).
Collapse
Affiliation(s)
- V L Lakomkin
- Chazov National Medical Research Center of Cardiology, Moscow
| | - A A Abramov
- Chazov National Medical Research Center of Cardiology, Moscow
| | - A V Prosvirnin
- Chazov National Medical Research Center of Cardiology, Moscow
| | | | - G K Arutunan
- Chazov National Medical Research Center of Cardiology, Moscow
| | - A N Samko
- Chazov National Medical Research Center of Cardiology, Moscow
| | - V I Kapelko
- Chazov National Medical Research Center of Cardiology, Moscow
| |
Collapse
|
14
|
Han YS, Pakkam M, Fogarty MJ, Sieck GC, Brozovich FV. Alterations in cardiac contractile and regulatory proteins contribute to age-related cardiac dysfunction in male rats. Physiol Rep 2024; 12:e70012. [PMID: 39169429 PMCID: PMC11338742 DOI: 10.14814/phy2.70012] [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: 03/26/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
Aging is associated with cardiac contractile abnormalities, but the etiology of these contractile deficits is unclear. We hypothesized that cardiac contractile and regulatory protein expression is altered during aging. To investigate this possibility, left ventricular (LV) lysates were prepared from young (6 months) and old (24 months) Fischer344 rats. There are no age-related changes in SERCA2 expression or phospholamban phosphorylation. Additionally, neither titin isoform expression nor phosphorylation differed. However, there is a significant increase in β-isoform of the myosin heavy chain (MyHC) expression and phosphorylation of TnI and MyBP-C during aging. In permeabilized strips of papillary muscle, force and Ca2+ sensitivity are reduced during aging, consistent with the increase in β-MyHC expression and TnI phosphorylation. However, the increase in MyBP-C phosphorylation during aging may represent a mechanism to compensate for age-related contractile deficits. In isolated cardiomyocytes loaded with Fura-2, the peak of the Ca2+ transient is reduced, but the kinetics of the Ca2+ transient are not altered. Furthermore, the extent of shortening and the rates of both sarcomere shortening and re-lengthening are reduced. These results demonstrate that aging is associated with changes in contractile and regulatory protein expression and phosphorylation, which affect the mechanical properties of cardiac muscle.
Collapse
Affiliation(s)
- Young Soo Han
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Madona Pakkam
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Matthew J. Fogarty
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Gary C. Sieck
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Frank V. Brozovich
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Department of Cardiovascular DiseasesMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
15
|
Douglas CM, Bird JE, Kopinke D, Esser KA. An optimized approach to study nanoscale sarcomere structure utilizing super-resolution microscopy with nanobodies. PLoS One 2024; 19:e0300348. [PMID: 38687705 PMCID: PMC11060602 DOI: 10.1371/journal.pone.0300348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/23/2024] [Indexed: 05/02/2024] Open
Abstract
The sarcomere is the fundamental contractile unit in skeletal muscle, and the regularity of its structure is critical for function. Emerging data demonstrates that nanoscale changes to the regularity of sarcomere structure can affect the overall function of the protein dense ~2μm sarcomere. Further, sarcomere structure is implicated in many clinical conditions of muscle weakness. However, our understanding of how sarcomere structure changes in disease, especially at the nanoscale, has been limited in part due to the inability to robustly detect and measure at sub-sarcomere resolution. We optimized several methodological steps and developed a robust pipeline to analyze sarcomere structure using structured illumination super-resolution microscopy in conjunction with commercially-available and fluorescently-conjugated Variable Heavy-Chain only fragment secondary antibodies (nanobodies), and achieved a significant increase in resolution of z-disc width (353nm vs. 62nm) compared to confocal microscopy. The combination of these methods provides a unique approach to probe sarcomere protein localization at the nanoscale and may prove advantageous for analysis of other cellular structures.
Collapse
Affiliation(s)
- Collin M. Douglas
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States of America
| | - Jonathan E. Bird
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Karyn A. Esser
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
16
|
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
|
17
|
Usui Y, Hanashima A, Hashimoto K, Kimoto M, Ohira M, Mohri S. Comparative analysis of ventricular stiffness across species. Physiol Rep 2024; 12:e16013. [PMID: 38644486 PMCID: PMC11033294 DOI: 10.14814/phy2.16013] [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: 09/20/2023] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 04/23/2024] Open
Abstract
Investigating ventricular diastolic properties is crucial for understanding the physiological cardiac functions in organisms and unraveling the pathological mechanisms of cardiovascular disorders. Ventricular stiffness, a fundamental parameter that defines ventricular diastolic functions in chordates, is typically analyzed using the end-diastolic pressure-volume relationship (EDPVR). However, comparing ventricular stiffness accurately across chambers of varying maximum volume capacities has been a long-standing challenge. As one of the solutions to this problem, we propose calculating a relative ventricular stiffness index by applying an exponential approximation formula to the EDPVR plot data of the relationship between ventricular pressure and values of normalized ventricular volume by the ventricular weight. This article reviews the potential, utility, and limitations of using normalized EDPVR analysis in recent studies. Herein, we measured and ranked ventricular stiffness in differently sized and shaped chambers using ex vivo ventricular pressure-volume analysis data from four animals: Wistar rats, red-eared slider turtles, masu salmon, and cherry salmon. Furthermore, we have discussed the mechanical effects of intracellular and extracellular viscoelastic components, Titin (Connectin) filaments, collagens, physiological sarcomere length, and other factors that govern ventricular stiffness. Our review provides insights into the comparison of ventricular stiffness in different-sized ventricles between heterologous and homologous species, including non-model organisms.
Collapse
Grants
- JP22K15155 Japan Society for the Promotion of Science, Grant/Award Number
- JP20K21453 Japan Society for the Promotion of Science, Grant/Award Number
- JP20H04508 Japan Society for the Promotion of Science, Grant/Award Number
- JP21K19933 Japan Society for the Promotion of Science, Grant/Award Number
- JP20H04521 Japan Society for the Promotion of Science, Grant/Award Number
- JP17H02092 Japan Society for the Promotion of Science, Grant/Award Number
- JP23H00556 Japan Society for the Promotion of Science, Grant/Award Number
- JP17H06272 Japan Society for the Promotion of Science, Grant/Award Number
- JP17H00859 Japan Society for the Promotion of Science, Grant/Award Number
- JP25560214 Japan Society for the Promotion of Science, Grant/Award Number
- JP16K01385 Japan Society for the Promotion of Science, Grant/Award Number
- JP26282127 Japan Society for the Promotion of Science, Grant/Award Number
- The Futaba research grant program
- Research Grant from the Kawasaki Foundation in 2016 from Medical Science and Medical Welfare
- Medical Research Grant in 2010 from Takeda Science Foundation
- R03S005 Research Project Grant from Kawasaki Medical School
- R03B050 Research Project Grant from Kawasaki Medical School
- R01B054 Research Project Grant from Kawasaki Medical School
- H30B041 Research Project Grant from Kawasaki Medical School
- H30B016 Research Project Grant from Kawasaki Medical School
- H27B10 Research Project Grant from Kawasaki Medical School
- R02B039 Research Project Grant from Kawasaki Medical School
- H28B80 Research Project Grant from Kawasaki Medical School
- R05B016 Research Project Grant from Kawasaki Medical School
- Japan Society for the Promotion of Science, Grant/Award Number
Collapse
Affiliation(s)
- Yuu Usui
- First Department of PhysiologyKawasaki Medical SchoolKurashikiOkayamaJapan
| | - Akira Hanashima
- First Department of PhysiologyKawasaki Medical SchoolKurashikiOkayamaJapan
| | - Ken Hashimoto
- First Department of PhysiologyKawasaki Medical SchoolKurashikiOkayamaJapan
| | - Misaki Kimoto
- First Department of PhysiologyKawasaki Medical SchoolKurashikiOkayamaJapan
| | - Momoko Ohira
- First Department of PhysiologyKawasaki Medical SchoolKurashikiOkayamaJapan
| | - Satoshi Mohri
- First Department of PhysiologyKawasaki Medical SchoolKurashikiOkayamaJapan
| |
Collapse
|
18
|
Hinson JT, Hershberger RE. Monoallelic TTN Truncation Variants Identified in Individuals With DCM May Cause a Mild Skeletal Myopathy. JACC. HEART FAILURE 2024; 12:754-756. [PMID: 38206233 DOI: 10.1016/j.jchf.2023.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 01/12/2024]
Affiliation(s)
- J Travis Hinson
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA; Cardiology Center, UConn Health, Farmington, Connecticut, USA.
| | - Ray E Hershberger
- Divisions of Human Genetics and Cardiovascular Medicine, Department of Medicine, The Ohio State University Medical Center, Columbus, Ohio, USA; The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, USA
| |
Collapse
|
19
|
Jolfayi AG, Kohansal E, Ghasemi S, Naderi N, Hesami M, MozafaryBazargany M, Moghadam MH, Fazelifar AF, Maleki M, Kalayinia S. Exploring TTN variants as genetic insights into cardiomyopathy pathogenesis and potential emerging clues to molecular mechanisms in cardiomyopathies. Sci Rep 2024; 14:5313. [PMID: 38438525 PMCID: PMC10912352 DOI: 10.1038/s41598-024-56154-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/01/2024] [Indexed: 03/06/2024] Open
Abstract
The giant protein titin (TTN) is a sarcomeric protein that forms the myofibrillar backbone for the components of the contractile machinery which plays a crucial role in muscle disorders and cardiomyopathies. Diagnosing TTN pathogenic variants has important implications for patient management and genetic counseling. Genetic testing for TTN variants can help identify individuals at risk for developing cardiomyopathies, allowing for early intervention and personalized treatment strategies. Furthermore, identifying TTN variants can inform prognosis and guide therapeutic decisions. Deciphering the intricate genotype-phenotype correlations between TTN variants and their pathologic traits in cardiomyopathies is imperative for gene-based diagnosis, risk assessment, and personalized clinical management. With the increasing use of next-generation sequencing (NGS), a high number of variants in the TTN gene have been detected in patients with cardiomyopathies. However, not all TTN variants detected in cardiomyopathy cohorts can be assumed to be disease-causing. The interpretation of TTN variants remains challenging due to high background population variation. This narrative review aimed to comprehensively summarize current evidence on TTN variants identified in published cardiomyopathy studies and determine which specific variants are likely pathogenic contributors to cardiomyopathy development.
Collapse
Affiliation(s)
- Amir Ghaffari Jolfayi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Erfan Kohansal
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Serwa Ghasemi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Naderi
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahshid Hesami
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Hosseini Moghadam
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Farjam Fazelifar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Kalayinia
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Janssens JV, Raaijmakers AJA, Weeks KL, Bell JR, Mellor KM, Curl CL, Delbridge LMD. The cardiomyocyte origins of diastolic dysfunction: cellular components of myocardial "stiffness". Am J Physiol Heart Circ Physiol 2024; 326:H584-H598. [PMID: 38180448 DOI: 10.1152/ajpheart.00334.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 12/07/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
The impaired ability of the heart to relax and stretch to accommodate venous return is generally understood to represent a state of "diastolic dysfunction" and often described using the all-purpose noun "stiffness." Despite the now common qualitative usage of this term in fields of cardiac patho/physiology, the specific quantitative concept of stiffness as a molecular and biophysical entity with real practical interpretation in healthy and diseased hearts is sometimes obscure. The focus of this review is to characterize the concept of cardiomyocyte stiffness and to develop interpretation of "stiffness" attributes at the cellular and molecular levels. Here, we consider "stiffness"-related terminology interpretation and make links between cardiomyocyte stiffness and aspects of functional and structural cardiac performance. We discuss cross bridge-derived stiffness sources, considering the contributions of diastolic myofilament activation and impaired relaxation. This includes commentary relating to the role of cardiomyocyte Ca2+ flux and Ca2+ levels in diastole, the troponin-tropomyosin complex role as a Ca2+ effector in diastole, the myosin ADP dissociation rate as a modulator of cross bridge attachment and regulation of cross-bridge attachment by myosin binding protein C. We also discuss non-cross bridge-derived stiffness sources, including the titin sarcomeric spring protein, microtubule and intermediate filaments, and cytoskeletal extracellular matrix interactions. As the prevalence of conditions involving diastolic heart failure has escalated, a more sophisticated understanding of the molecular, cellular, and tissue determinants of cardiomyocyte stiffness offers potential to develop imaging and molecular intervention tools.
Collapse
Affiliation(s)
- Johannes V Janssens
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Antonia J A Raaijmakers
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Kate L Weeks
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
- Department of Diabetes, Monash University, Parkville, Victoria, Australia
| | - James R Bell
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - Kimberley M Mellor
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Physiology, University of Auckland, Auckland, New Zealand
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Claire L Curl
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Lea M D Delbridge
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Weston TGR, Rees M, Gautel M, Fraternali F. Walking with giants: The challenges of variant impact assessment in the giant sarcomeric protein titin. WIREs Mech Dis 2024; 16:e1638. [PMID: 38155593 DOI: 10.1002/wsbm.1638] [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: 06/15/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
Titin, the so-called "third filament" of the sarcomere, represents a difficult challenge for the determination of damaging genetic variants. A single titin molecule extends across half the length of a sarcomere in striated muscle, fulfilling a variety of vital structural and signaling roles, and has been linked to an equally varied range of myopathies, resulting in a significant burden on individuals and healthcare systems alike. While the consequences of truncating variants of titin are well-documented, the ramifications of the missense variants prevalent in the general population are less so. We here present a compendium of titin missense variants-those that result in a single amino-acid substitution in coding regions-reported to be pathogenic and discuss these in light of the nature of titin and the variant position within the sarcomere and their domain, the structural, pathological, and biophysical characteristics that define them, and the methods used for characterization. Finally, we discuss the current knowledge and integration of the multiple fields that have contributed to our understanding of titin-related pathology and offer suggestions as to how these concurrent methodologies may aid the further development in our understanding of titin and hopefully extend to other, less well-studied giant proteins. This article is categorized under: Cardiovascular Diseases > Genetics/Genomics/Epigenetics Congenital Diseases > Genetics/Genomics/Epigenetics Congenital Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Timir G R Weston
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Martin Rees
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Mathias Gautel
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Franca Fraternali
- Institute of Structural and Molecular Biology, University College London, London, UK
| |
Collapse
|
22
|
Ma W, Lee KH, Delligatti CE, Davis MT, Zheng Y, Gong H, Kirk JA, Craig R, Irving T. The structural and functional integrities of porcine myocardium are mostly preserved by cryopreservation. J Gen Physiol 2023; 155:e202313345. [PMID: 37398997 PMCID: PMC10318404 DOI: 10.1085/jgp.202313345] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/05/2023] [Accepted: 06/21/2023] [Indexed: 07/04/2023] Open
Abstract
Structural and functional studies of heart muscle are important to gain insights into the physiological bases of cardiac muscle contraction and the pathological bases of heart disease. While fresh muscle tissue works best for these kinds of studies, this is not always practical to obtain, especially for heart tissue from large animal models and humans. Conversely, tissue banks of frozen human hearts are available and could be a tremendous resource for translational research. It is not well understood, however, how liquid nitrogen freezing and cryostorage may impact the structural integrity of myocardium from large mammals. In this study, we directly compared the structural and functional integrity of never-frozen to previously frozen porcine myocardium to investigate the consequences of freezing and cryostorage. X-ray diffraction measurements from hydrated tissue under near-physiological conditions and electron microscope images from chemically fixed porcine myocardium showed that prior freezing has only minor effects on structural integrity of the muscle. Furthermore, mechanical studies similarly showed no significant differences in contractile capabilities of porcine myocardium with and without freezing and cryostorage. These results demonstrate that liquid nitrogen preservation is a practical approach for structural and functional studies of myocardium.
Collapse
Affiliation(s)
- Weikang Ma
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Kyoung Hwan Lee
- Electron Microscopy Facility, UMass Chan Medical School, Worcester, MA, USA
| | | | - M. Therese Davis
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, IL, USA
| | - Yahan Zheng
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Henry Gong
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Jonathan A. Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, IL, USA
| | - Roger Craig
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Thomas Irving
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| |
Collapse
|
23
|
Li J, Sundnes J, Hou Y, Laasmaa M, Ruud M, Unger A, Kolstad TR, Frisk M, Norseng PA, Yang L, Setterberg IE, Alves ES, Kalakoutis M, Sejersted OM, Lanner JT, Linke WA, Lunde IG, de Tombe PP, Louch WE. Stretch Harmonizes Sarcomere Strain Across the Cardiomyocyte. Circ Res 2023; 133:255-270. [PMID: 37401464 PMCID: PMC10355805 DOI: 10.1161/circresaha.123.322588] [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: 01/27/2023] [Revised: 06/07/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND Increasing cardiomyocyte contraction during myocardial stretch serves as the basis for the Frank-Starling mechanism in the heart. However, it remains unclear how this phenomenon occurs regionally within cardiomyocytes, at the level of individual sarcomeres. We investigated sarcomere contractile synchrony and how intersarcomere dynamics contribute to increasing contractility during cell lengthening. METHODS Sarcomere strain and Ca2+ were simultaneously recorded in isolated left ventricular cardiomyocytes during 1 Hz field stimulation at 37 °C, at resting length and following stepwise stretch. RESULTS We observed that in unstretched rat cardiomyocytes, differential sarcomere deformation occurred during each beat. Specifically, while most sarcomeres shortened during the stimulus, ≈10% to 20% of sarcomeres were stretched or remained stationary. This nonuniform strain was not traced to regional Ca2+ disparities but rather shorter resting lengths and lower force production in systolically stretched sarcomeres. Lengthening of the cell recruited additional shortening sarcomeres, which increased contractile efficiency as less negative, wasted work was performed by stretched sarcomeres. Given the known role of titin in setting sarcomere dimensions, we next hypothesized that modulating titin expression would alter intersarcomere dynamics. Indeed, in cardiomyocytes from mice with titin haploinsufficiency, we observed greater variability in resting sarcomere length, lower recruitment of shortening sarcomeres, and impaired work performance during cell lengthening. CONCLUSIONS Graded sarcomere recruitment directs cardiomyocyte work performance, and harmonization of sarcomere strain increases contractility during cell stretch. By setting sarcomere dimensions, titin controls sarcomere recruitment, and its lowered expression in haploinsufficiency mutations impairs cardiomyocyte contractility.
Collapse
Affiliation(s)
- Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | | | - Yufeng Hou
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Martin Laasmaa
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Marianne Ruud
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Andreas Unger
- Institute of Physiology II, University of Münster, Germany (A.U., W.A.L.)
| | - Terje R. Kolstad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Per Andreas Norseng
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
| | | | - Ingunn E. Setterberg
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Estela S. Alves
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (E.S.A., M.K., J.T.L.)
| | - Michaeljohn Kalakoutis
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (E.S.A., M.K., J.T.L.)
| | - Ole M. Sejersted
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Johanna T. Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (E.S.A., M.K., J.T.L.)
| | - Wolfgang A. Linke
- Institute of Physiology II, University of Münster, Germany (A.U., W.A.L.)
| | - Ida G. Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Pieter P. de Tombe
- Department of Physiology and Biophysics, University of Illinois at Chicago (P.P.d.T.)
- Phymedexp, Université de Montpellier, INSERM, CNRS, France (P.P.d.T.)
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| |
Collapse
|
24
|
Linke WA. Stretching the story of titin and muscle function. J Biomech 2023; 152:111553. [PMID: 36989971 DOI: 10.1016/j.jbiomech.2023.111553] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023]
Abstract
The discovery of the giant protein titin, also known as connectin, dates almost half a century back. In this review, I recapitulate major advances in the discovery of the titin filaments and the recognition of their properties and function until today. I briefly discuss how our understanding of the layout and interactions of titin in muscle sarcomeres has evolved and review key facts about the titin sequence at the gene (TTN) and protein levels. I also touch upon properties of titin important for the stability of the contractile units and the assembly and maintenance of sarcomeric proteins. The greater part of my discussion centers around the mechanical function of titin in skeletal muscle. I cover milestones of research on titin's role in stretch-dependent passive tension development, recollect the reasons behind the enormous elastic diversity of titin, and provide an update on the molecular mechanisms of titin elasticity, details of which are emerging even now. I reflect on current knowledge of how muscle fibers behave mechanically if titin stiffness is removed and how titin stiffness can be dynamically regulated, such as by posttranslational modifications or calcium binding. Finally, I highlight novel and exciting, but still controversially discussed, insight into the role titin plays in active tension development, such as length-dependent activation and contraction from longer muscle lengths.
Collapse
Affiliation(s)
- Wolfgang A Linke
- Institute of Physiology II, University of Münster, Germany; Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Germany; German Centre for Cardiovascular Research, Berlin, Germany.
| |
Collapse
|
25
|
Han SW, Boldt K, Joumaa V, Herzog W. Characterizing residual and passive force enhancements in cardiac myofibrils. Biophys J 2023; 122:1538-1547. [PMID: 36932677 PMCID: PMC10147830 DOI: 10.1016/j.bpj.2023.03.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 11/07/2022] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Residual force enhancement (RFE), an increase in isometric force after active stretching of a muscle compared with the purely isometric force at the corresponding length, has been consistently observed throughout the structural hierarchy of skeletal muscle. Similar to RFE, passive force enhancement (PFE) is also observable in skeletal muscle and is defined as an increase in passive force when a muscle is deactivated after it has been actively stretched compared with the passive force following deactivation of a purely isometric contraction. These history-dependent properties have been investigated abundantly in skeletal muscle, but their presence in cardiac muscle remains unresolved and controversial. The purpose of this study was to investigate whether RFE and PFE exist in cardiac myofibrils and whether the magnitudes of RFE and PFE increase with increasing stretch magnitudes. Cardiac myofibrils were prepared from the left ventricles of New Zealand White rabbits, and the history-dependent properties were tested at three different final average sarcomere lengths (n = 8 for each), 1.8, 2, and 2.2 μm, while the stretch magnitude was kept at 0.2 μm/sarcomere. The same experiment was repeated with a final average sarcomere length of 2.2 μm and a stretching magnitude of 0.4 μm/sarcomere (n = 8). All 32 cardiac myofibrils exhibited increased forces after active stretching compared with the corresponding purely isometric reference conditions (p < 0.05). Furthermore, the magnitude of RFE was greater when myofibrils were stretched by 0.4 compared with 0.2 μm/sarcomere (p < 0.05). We conclude that, like in skeletal muscle, RFE and PFE are properties of cardiac myofibrils and are dependent on stretch magnitude.
Collapse
Affiliation(s)
- Seong-Won Han
- Institute of Physiology II, Faculty of Medicine, University of Münster, Münster, Germany; Faculty of Kinesiology, University of Calgary, Calgary, Canada.
| | - Kevin Boldt
- Faculty of Kinesiology, University of Calgary, Calgary, Canada; Kinesiology Program, Trent University, Peterborough, ON, Canada; Department of Human Health and Nutritional Science, University of Guelph, Guelph, ON, Canada
| | - Venus Joumaa
- Faculty of Kinesiology, University of Calgary, Calgary, Canada
| | - Walter Herzog
- Faculty of Kinesiology, University of Calgary, Calgary, Canada
| |
Collapse
|
26
|
Feng HZ, Huang X, Jin JP. N-terminal truncated cardiac troponin I enhances Frank-Starling response by increasing myofilament sensitivity to resting tension. J Gen Physiol 2023; 155:e202012821. [PMID: 36880803 PMCID: PMC10005897 DOI: 10.1085/jgp.202012821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/09/2022] [Accepted: 12/29/2022] [Indexed: 03/08/2023] Open
Abstract
Cardiac troponin I (cTnI) of higher vertebrates has evolved with an N-terminal extension, of which deletion via restrictive proteolysis occurs as a compensatory adaptation in chronic heart failure to increase ventricular relaxation and stroke volume. Here, we demonstrate in a transgenic mouse model expressing solely N-terminal truncated cTnI (cTnI-ND) in the heart with deletion of the endogenous cTnI gene. Functional studies using ex vivo working hearts showed an extended Frank-Starling response to preload with reduced left ventricular end diastolic pressure. The enhanced Frank-Starling response effectively increases systolic ventricular pressure development and stroke volume. A novel finding is that cTnI-ND increases left ventricular relaxation velocity and stroke volume without increasing the end diastolic volume. Consistently, the optimal resting sarcomere length (SL) for maximum force development in cTnI-ND cardiac muscle was not different from wild-type (WT) control. Despite the removal of the protein kinase A (PKA) phosphorylation sites in cTnI, β-adrenergic stimulation remains effective on augmenting the enhanced Frank-Starling response of cTnI-ND hearts. Force-pCa relationship studies using skinned preparations found that while cTnI-ND cardiac muscle shows a resting SL-resting tension relationship similar to WT control, cTnI-ND significantly increases myofibril Ca2+ sensitivity to resting tension. The results demonstrate that restrictive N-terminal deletion of cTnI enhances Frank-Starling response by increasing myofilament sensitivity to resting tension rather than directly depending on SL. This novel function of cTnI regulation suggests a myofilament approach to utilizing Frank-Starling mechanism for the treatment of heart failure, especially diastolic failure where ventricular filling is limited.
Collapse
Affiliation(s)
- Han-Zhong Feng
- Department of Physiology and Biophysics, University of Illinois at Chicago School of Medicine, Chicago, IL, USA
| | - Xupei Huang
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Jian-Ping Jin
- Department of Physiology and Biophysics, University of Illinois at Chicago School of Medicine, Chicago, IL, USA
| |
Collapse
|
27
|
Tanner BCW, Awinda PO, Agonias KB, Attili S, Blair CA, Thompson MS, Walker LA, Kampourakis T, Campbell KS. Sarcomere length affects Ca2+ sensitivity of contraction in ischemic but not non-ischemic myocardium. J Gen Physiol 2023; 155:213800. [PMID: 36633584 PMCID: PMC9859763 DOI: 10.1085/jgp.202213200] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/18/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
In healthy hearts, myofilaments become more sensitive to Ca2+ as the myocardium is stretched. This effect is known as length-dependent activation and is an important cellular-level component of the Frank-Starling mechanism. Few studies have measured length-dependent activation in the myocardium from failing human hearts. We investigated whether ischemic and non-ischemic heart failure results in different length-dependent activation responses at physiological temperature (37°C). Myocardial strips from the left ventricular free wall were chemically permeabilized and Ca2+-activated at sarcomere lengths (SLs) of 1.9 and 2.3 µm. Data were acquired from 12 hearts that were explanted from patients receiving cardiac transplants; 6 had ischemic heart failure and 6 had non-ischemic heart failure. Another 6 hearts were obtained from organ donors. Maximal Ca2+-activated force increased at longer SL for all groups. Ca2+ sensitivity increased with SL in samples from donors (P < 0.001) and patients with ischemic heart failure (P = 0.003) but did not change with SL in samples from patients with non-ischemic heart failure. Compared with donors, troponin I phosphorylation decreased in ischemic samples and even more so in non-ischemic samples; cardiac myosin binding protein-C (cMyBP-C) phosphorylation also decreased with heart failure. These findings support the idea that troponin I and cMyBP-C phosphorylation promote length-dependent activation and show that length-dependent activation of contraction is blunted, yet extant, in the myocardium from patients with ischemic heart failure and further reduced in the myocardium from patients with non-ischemic heart failure. Patients who have a non-ischemic disease may exhibit a diminished contractile response to increased ventricular filling.
Collapse
Affiliation(s)
- Bertrand C W Tanner
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, WA, USA
| | - Peter O Awinda
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, WA, USA
| | - Keinan B Agonias
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, WA, USA
| | - Seetharamaiah Attili
- Randall Centre for Cell and Molecular Biophysics, King's College London , London, UK
| | - Cheavar A Blair
- Department of Physiology, University of Kentucky , Lexington, KY, USA
| | - Mindy S Thompson
- Department of Physiology, University of Kentucky , Lexington, KY, USA
| | - Lori A Walker
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus , Aurora, CO, USA
| | - Thomas Kampourakis
- Randall Centre for Cell and Molecular Biophysics, King's College London , London, UK
| | - Kenneth S Campbell
- Department of Physiology, University of Kentucky , Lexington, KY, USA.,Division of Cardiovascular Medicine, University of Kentucky , Lexington, KY, USA
| |
Collapse
|
28
|
Chen Y, Ma T, Zhang T, Ma L. Trends in the evolution of intronless genes in Poaceae. FRONTIERS IN PLANT SCIENCE 2023; 14:1065631. [PMID: 36875616 PMCID: PMC9978806 DOI: 10.3389/fpls.2023.1065631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Intronless genes (IGs), which are a feature of prokaryotes, are a fascinating group of genes that are also present in eukaryotes. In the current study, a comparison of Poaceae genomes revealed that the origin of IGs may have involved ancient intronic splicing, reverse transcription, and retrotranspositions. Additionally, IGs exhibit the typical features of rapid evolution, including recent duplications, variable copy numbers, low divergence between paralogs, and high non-synonymous to synonymous substitution ratios. By tracing IG families along the phylogenetic tree, we determined that the evolutionary dynamics of IGs differed among Poaceae subfamilies. IG families developed rapidly before the divergence of Pooideae and Oryzoideae and expanded slowly after the divergence. In contrast, they emerged gradually and consistently in the Chloridoideae and Panicoideae clades during evolution. Furthermore, IGs are expressed at low levels. Under relaxed selection pressure, retrotranspositions, intron loss, and gene duplications and conversions may promote the evolution of IGs. The comprehensive characterization of IGs is critical for in-depth studies on intron functions and evolution as well as for assessing the importance of introns in eukaryotes.
Collapse
Affiliation(s)
- Yong Chen
- *Correspondence: Tingting Zhang, ; Lei Ma,
| | | | | | - Lei Ma
- *Correspondence: Tingting Zhang, ; Lei Ma,
| |
Collapse
|
29
|
Vintrych P, Al-Obeidallah M, Horák J, Chvojka J, Valešová L, Nalos L, Jarkovská D, Matějovič M, Štengl M. Modeling sepsis, with a special focus on large animal models of porcine peritonitis and bacteremia. Front Physiol 2023; 13:1094199. [PMID: 36703923 PMCID: PMC9871395 DOI: 10.3389/fphys.2022.1094199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Infectious diseases, which often result in deadly sepsis or septic shock, represent a major global health problem. For understanding the pathophysiology of sepsis and developing new treatment strategies, reliable and clinically relevant animal models of the disease are necessary. In this review, two large animal (porcine) models of sepsis induced by either peritonitis or bacteremia are introduced and their strong and weak points are discussed in the context of clinical relevance and other animal models of sepsis, with a special focus on cardiovascular and immune systems, experimental design, and monitoring. Especially for testing new therapeutic strategies, the large animal (porcine) models represent a more clinically relevant alternative to small animal models, and the findings obtained in small animal (transgenic) models should be verified in these clinically relevant large animal models before translation to the clinical level.
Collapse
Affiliation(s)
- Pavel Vintrych
- Department of Cardiology, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Mahmoud Al-Obeidallah
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Jan Horák
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Jiří Chvojka
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Lenka Valešová
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Lukáš Nalos
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Dagmar Jarkovská
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Martin Matějovič
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Milan Štengl
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,*Correspondence: Milan Štengl,
| |
Collapse
|
30
|
Sy MR, Keefe JA, Sutton JP, Wehrens XHT. Cardiac function, structural, and electrical remodeling by microgravity exposure. Am J Physiol Heart Circ Physiol 2023; 324:H1-H13. [PMID: 36399385 PMCID: PMC9762974 DOI: 10.1152/ajpheart.00611.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
Space medicine is key to the human exploration of outer space and pushes the boundaries of science, technology, and medicine. Because of harsh environmental conditions related to microgravity and other factors and hazards in outer space, astronauts and spaceflight participants face unique health and medical challenges, including those related to the heart. In this review, we summarize the literature regarding the effects of spaceflight on cardiac structure and function. We also provide an in-depth review of the literature regarding the effects of microgravity on cardiac calcium handling. Our review can inform future mechanistic and therapeutic studies and is applicable to other physiological states similar to microgravity such as prolonged horizontal bed rest and immobilization.
Collapse
Affiliation(s)
- Mary R Sy
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas
| | - Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas
| | - Jeffrey P Sutton
- Center for Space Medicine, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas
- Center for Space Medicine, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
31
|
Ahmed RE, Tokuyama T, Anzai T, Chanthra N, Uosaki H. Sarcomere maturation: function acquisition, molecular mechanism, and interplay with other organelles. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210325. [PMID: 36189811 PMCID: PMC9527934 DOI: 10.1098/rstb.2021.0325] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 06/15/2022] [Indexed: 12/31/2022] Open
Abstract
During postnatal cardiac development, cardiomyocytes mature and turn into adult ones. Hence, all cellular properties, including morphology, structure, physiology and metabolism, are changed. One of the most important aspects is the contractile apparatus, of which the minimum unit is known as a sarcomere. Sarcomere maturation is evident by enhanced sarcomere alignment, ultrastructural organization and myofibrillar isoform switching. Any maturation process failure may result in cardiomyopathy. Sarcomere function is intricately related to other organelles, and the growing evidence suggests reciprocal regulation of sarcomere and mitochondria on their maturation. Herein, we summarize the molecular mechanism that regulates sarcomere maturation and the interplay between sarcomere and other organelles in cardiomyocyte maturation. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- Razan E. Ahmed
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Takeshi Tokuyama
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Tatsuya Anzai
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Nawin Chanthra
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Hideki Uosaki
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
32
|
Usui Y, Kimoto M, Hanashima A, Hashimoto K, Mohri S. Cardiac hemodynamics and ventricular stiffness of sea-run cherry salmon (Oncorhynchus masou masou) differ critically from those of landlocked masu salmon. PLoS One 2022; 17:e0267264. [PMID: 36331913 PMCID: PMC9635730 DOI: 10.1371/journal.pone.0267264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Ventricular diastolic mechanical properties are important determinants of cardiac function and are optimized by changes in cardiac structure and physical properties. Oncorhynchus masou masou is an anadromous migratory fish of the Salmonidae family, and several ecological studies on it have been conducted; however, the cardiac functions of the fish are not well known. Therefore, we investigated ventricular diastolic function in landlocked (masu salmon) and sea-run (cherry salmon) types at 29–30 months post fertilization. Pulsed-wave Doppler echocardiography showed that the atrioventricular inflow waveforms of cherry salmon were biphasic with early diastolic filling and atrial contraction, whereas those of masu salmon were monophasic with atrial contraction. In addition, end-diastolic pressure–volume relationship analysis revealed that the dilatability per unit myocardial mass of the ventricle in cherry salmon was significantly suppressed compared to that in masu salmon, suggesting that the ventricle of the cherry salmon was relatively stiffer (relative ventricular stiffness index; p = 0.0263). Contrastingly, the extensibility of cardiomyocytes, characterized by the expression pattern of Connectin isoforms in their ventricles, was similar in both types. Histological analysis showed that the percentage of the collagen accumulation area in the compact layer of cherry salmon increased compared with that of the masu salmon, which may contribute to ventricle stiffness. Although the heart mass of cherry salmon was about 11-fold greater than that of masu salmon, there was no difference in the morphology of the isolated cardiomyocytes, suggesting that the heart of the cherry salmon grows by cardiomyocyte proliferation, but not cell hypertrophy. The cardiac physiological function of the teleosts varies with differences in their developmental processes and life history. Our multidimensional analysis of the O. masou heart may provide a clue to the process by which the heart acquires a biphasic blood-filling pattern, i.e., a ventricular diastolic suction.
Collapse
Affiliation(s)
- Yuu Usui
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Japan
- * E-mail:
| | - Misaki Kimoto
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Japan
| | - Akira Hanashima
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Japan
| | - Ken Hashimoto
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Japan
| | - Satoshi Mohri
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Japan
| |
Collapse
|
33
|
Herrero-Galán E, Martínez-Martín I, Sánchez-González C, Vicente N, Bonzón-Kulichenko E, Calvo E, Suay-Corredera C, Pricolo MR, Fernández-Trasancos Á, Velázquez-Carreras D, Careaga CB, Abdellatif M, Sedej S, Rainer PP, Giganti D, Pérez-Jiménez R, Vázquez J, Alegre-Cebollada J. Basal oxidation of conserved cysteines modulates cardiac titin stiffness and dynamics. Redox Biol 2022; 52:102306. [PMID: 35367810 PMCID: PMC8971355 DOI: 10.1016/j.redox.2022.102306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 01/11/2023] Open
Abstract
Titin, as the main protein responsible for the passive stiffness of the sarcomere, plays a key role in diastolic function and is a determinant factor in the etiology of heart disease. Titin stiffness depends on unfolding and folding transitions of immunoglobulin-like (Ig) domains of the I-band, and recent studies have shown that oxidative modifications of cryptic cysteines belonging to these Ig domains modulate their mechanical properties in vitro. However, the relevance of this mode of titin mechanical modulation in vivo remains largely unknown. Here, we describe the high evolutionary conservation of titin mechanical cysteines and show that they are remarkably oxidized in murine cardiac tissue. Mass spectrometry analyses indicate a similar landscape of basal oxidation in murine and human myocardium. Monte Carlo simulations illustrate how disulfides and S-thiolations on these cysteines increase the dynamics of the protein at physiological forces, while enabling load- and isoform-dependent regulation of titin stiffness. Our results demonstrate the role of conserved cysteines in the modulation of titin mechanical properties in vivo and point to potential redox-based pathomechanisms in heart disease.
Collapse
Affiliation(s)
| | | | | | - Natalia Vicente
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Elena Bonzón-Kulichenko
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Enrique Calvo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | | | | | | | | | | | - Simon Sedej
- Division of Cardiology, Medical University of Graz, Graz, Austria; Faculty of Medicine, University of Maribor, Maribor, Slovenia; BioTechMed Graz, Graz, Austria
| | - Peter P Rainer
- Division of Cardiology, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria
| | - David Giganti
- Department of Biochemistry & Molecular Pharmacology and Institute for Systems Genetics, NYU Langone Health, New York, NY, United States
| | - Raúl Pérez-Jiménez
- CIC NanoGUNE BRTA, San Sebastian, Spain; Ikerbasque Foundation for Science, Bilbao, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | |
Collapse
|
34
|
Baumert P, Cocks M, Strauss JA, Shepherd SO, Drust B, Lake MJ, Stewart CE, Erskine RM. Polygenic mechanisms underpinning the response to exercise‐induced muscle damage in humans: In vivo and in vitro evidence. J Cell Physiol 2022; 237:2862-2876. [DOI: 10.1002/jcp.30723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/21/2022] [Accepted: 03/07/2022] [Indexed: 11/12/2022]
Affiliation(s)
- Philipp Baumert
- Exercise Biology Group, Faculty of Sport and Health Sciences Technical University of Munich Munich Germany
- School of Sport and Exercise Sciences Liverpool John Moores University Liverpool UK
| | - Matthew Cocks
- School of Sport and Exercise Sciences Liverpool John Moores University Liverpool UK
| | - Juliette A. Strauss
- School of Sport and Exercise Sciences Liverpool John Moores University Liverpool UK
| | - Sam O. Shepherd
- School of Sport and Exercise Sciences Liverpool John Moores University Liverpool UK
| | - Barry Drust
- School of Sport, Exercise and Rehabilitation Sciences University of Birmingham Birmingham UK
| | - Mark J. Lake
- School of Sport and Exercise Sciences Liverpool John Moores University Liverpool UK
| | - Claire E. Stewart
- School of Sport and Exercise Sciences Liverpool John Moores University Liverpool UK
| | - Robert M. Erskine
- Institute of Sport, Exercise and Health University College London London UK
| |
Collapse
|
35
|
The Oxidative Balance Orchestrates the Main Keystones of the Functional Activity of Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7714542. [PMID: 35047109 PMCID: PMC8763515 DOI: 10.1155/2022/7714542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/03/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
This review is aimed at providing an overview of the key hallmarks of cardiomyocytes in physiological and pathological conditions. The main feature of cardiac tissue is the force generation through contraction. This process requires a conspicuous energy demand and therefore an active metabolism. The cardiac tissue is rich of mitochondria, the powerhouses in cells. These organelles, producing ATP, are also the main sources of ROS whose altered handling can cause their accumulation and therefore triggers detrimental effects on mitochondria themselves and other cell components thus leading to apoptosis and cardiac diseases. This review highlights the metabolic aspects of cardiomyocytes and wanders through the main systems of these cells: (a) the unique structural organization (such as different protein complexes represented by contractile, regulatory, and structural proteins); (b) the homeostasis of intracellular Ca2+ that represents a crucial ion for cardiac functions and E-C coupling; and (c) the balance of Zn2+, an ion with a crucial impact on the cardiovascular system. Although each system seems to be independent and finely controlled, the contractile proteins, intracellular Ca2+ homeostasis, and intracellular Zn2+ signals are strongly linked to each other by the intracellular ROS management in a fascinating way to form a "functional tetrad" which ensures the proper functioning of the myocardium. Nevertheless, if ROS balance is not properly handled, one or more of these components could be altered resulting in deleterious effects leading to an unbalance of this "tetrad" and promoting cardiovascular diseases. In conclusion, this "functional tetrad" is proposed as a complex network that communicates continuously in the cardiomyocytes and can drive the switch from physiological to pathological conditions in the heart.
Collapse
|
36
|
Corporan D, Segura A, Padala M. Ultrastructural Adaptation of the Cardiomyocyte to Chronic Mitral Regurgitation. Front Cardiovasc Med 2021; 8:714774. [PMID: 34733889 PMCID: PMC8559873 DOI: 10.3389/fcvm.2021.714774] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/14/2021] [Indexed: 01/18/2023] Open
Abstract
Introduction: Mitral regurgitation (MR) imposes volume overload on the left ventricle (LV) and elevates wall stress, triggering its adverse remodeling. Pronounced LV dilation, minimal wall thinning, and a gradual decline in cardiac ejection fraction (EF) are observed. The structural changes in the myocardium that define these gross, organ level remodeling are not known. Cardiomyocyte elongation and slippage have both been hypothesized, but neither are confirmed, nor are the changes to the cardiomyocyte structure known. Using a rodent model of MR, we used immunohistochemistry and transmission electron microscopy (TEM) to describe the ultrastructural remodeling of the cardiomyocyte. Methods: Twenty-four male Sprague-Dawley rats (350–400 g) were assigned to two groups: group (1) rats induced with severe MR (n = 18) and group (2) control rats that were healthy and age and weight matched (n = 6). MR was induced in the beating heart using a 23-G ultrasound-guided, transapical needle to perforate the anterior mitral leaflet, and the rats were followed to 2, 10, and 20 weeks (n = 6/time-point). Echocardiography was performed to quantify MR severity and to measure LV volume and function at each time-point. Explanted myocardial tissue were examined with TEM and immunohistochemistry to investigate the ultrastructural changes. Results: MR induced rapid and significant increase in end-diastolic volume (EDV), with a 50% increase by 2 weeks, compared with control. Rise in end-systolic volume (ESV) was more gradual; however, by 20 weeks, both EDV and ESV in MR rats were increased by 126% compared with control. A significant decline in EF was measured at 10 weeks of MR. At the ultrastructural level, as early as 2 weeks after MR, cardiomyocyte elongation and increase in cross-sectional area were observed. TEM depicted sarcomere shortening, with loss of Z-line and I-band. Desmin, a cytoskeletal protein that is uniformly distributed along the length of the cardiomyocyte, was disorganized and localized to the intercalated disc, in the rats induced with MR and not in the controls. In the rats with MR, the linear registry of the mitochondrial arrangement along the sarcomeres was lost, with mitochondrial fragmentation, aggregation around the nucleus, and irregularities in the cristae. Discussion: In the setting of chronic mitral regurgitation, LV dilatation occured by cardiomyocyte elongation, which manifests at the subcellular level as distinct ultrastructural alterations of the sarcomere, cytoskeleton, and mitochondria. Since the cytoskeleton not only provides tensegrity but has functional consequences on myocyte function, further investigation into the impact of cytoskeletal remodeling on progressive heart failure or recovery of function upon correcting the valve lesion are needed.
Collapse
Affiliation(s)
- Daniella Corporan
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center, Emory University Hospital Midtown, Atlanta, GE, United States.,Division of Cardiothoracic Surgery, Department of Surgery, School of Medicine, Emory University, Atlanta, GE, United States
| | - Ana Segura
- Department of Pathology, Texas Heart Institute, Houston, TX, United States
| | - Muralidhar Padala
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center, Emory University Hospital Midtown, Atlanta, GE, United States.,Division of Cardiothoracic Surgery, Department of Surgery, School of Medicine, Emory University, Atlanta, GE, United States
| |
Collapse
|
37
|
Kobirumaki-Shimozawa F, Shimozawa T, Oyama K, Baba S, Li J, Nakanishi T, Terui T, Louch WE, Ishiwata S, Fukuda N. Synchrony of sarcomeric movement regulates left ventricular pump function in the in vivo beating mouse heart. J Gen Physiol 2021; 153:212675. [PMID: 34605861 PMCID: PMC8493835 DOI: 10.1085/jgp.202012860] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 09/03/2021] [Indexed: 11/20/2022] Open
Abstract
Sarcomeric contraction in cardiomyocytes serves as the basis for the heart's pump functions. It has generally been considered that in cardiac muscle as well as in skeletal muscle, sarcomeres equally contribute to myofibrillar dynamics in myocytes at varying loads by producing similar levels of active and passive force. In the present study, we expressed α-actinin-AcGFP in Z-disks to analyze dynamic behaviors of sequentially connected individual sarcomeres along a myofibril in a left ventricular (LV) myocyte of the in vivo beating mouse heart. To quantify the magnitude of the contribution of individual sarcomeres to myofibrillar dynamics, we introduced the novel parameter "contribution index" (CI) to measure the synchrony in movements between a sarcomere and a myofibril (from -1 [complete asynchrony] to 1 [complete synchrony]). First, CI varied markedly between sarcomeres, with an average value of ∼0.3 during normal systole. Second, when the movements between adjacent sarcomeres were asynchronous (CI < 0), a sarcomere and the ones next to the adjacent sarcomeres and farther away moved in synchrony (CI > 0) along a myofibril. Third, when difference in LV pressure in diastole and systole (ΔLVP) was lowered to <10 mm Hg, diastolic sarcomere length increased. Under depressed conditions, the movements between adjacent sarcomeres were in marked asynchrony (CI, -0.3 to -0.4), and, as a result, average CI was linearly decreased in association with a decrease in ΔLVP. These findings suggest that in the left ventricle of the in vivo beating mouse heart, (1) sarcomeres heterogeneously contribute to myofibrillar dynamics due to an imbalance of active and passive force between neighboring sarcomeres, (2) the force imbalance is pronounced under depressed conditions coupled with a marked increase in passive force and the ensuing tug-of-war between sarcomeres, and (3) sarcomere synchrony via the distal intersarcomere interaction regulates the heart's pump function in coordination with myofibrillar contractility.
Collapse
Affiliation(s)
| | - Togo Shimozawa
- Technical Division, School of Science, The University of Tokyo, Tokyo, Japan
| | - Kotaro Oyama
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan.,Quantum Beam Science Research Directorate, National Institutes for Quantum and Radiological Science and Technology, Gunma, Japan
| | - Shunsuke Baba
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Tomohiro Nakanishi
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takako Terui
- Department of Anesthesiology, The Jikei University School of Medicine, Tokyo, Japan
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,K.G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Shin'ichi Ishiwata
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo, Japan
| | - Norio Fukuda
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
38
|
Sharifi H, Mann CK, Rockward AL, Mehri M, Mojumder J, Lee LC, Campbell KS, Wenk JF. Multiscale simulations of left ventricular growth and remodeling. Biophys Rev 2021; 13:729-746. [PMID: 34777616 PMCID: PMC8555068 DOI: 10.1007/s12551-021-00826-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiomyocytes can adapt their size, shape, and orientation in response to altered biomechanical or biochemical stimuli. The process by which the heart undergoes structural changes-affecting both geometry and material properties-in response to altered ventricular loading, altered hormonal levels, or mutant sarcomeric proteins is broadly known as cardiac growth and remodeling (G&R). Although it is likely that cardiac G&R initially occurs as an adaptive response of the heart to the underlying stimuli, prolonged pathological changes can lead to increased risk of atrial fibrillation, heart failure, and sudden death. During the past few decades, computational models have been extensively used to investigate the mechanisms of cardiac G&R, as a complement to experimental measurements. These models have provided an opportunity to quantitatively study the relationships between the underlying stimuli (primarily mechanical) and the adverse outcomes of cardiac G&R, i.e., alterations in ventricular size and function. State-of-the-art computational models have shown promise in predicting the progression of cardiac G&R. However, there are still limitations that need to be addressed in future works to advance the field. In this review, we first outline the current state of computational models of cardiac growth and myofiber remodeling. Then, we discuss the potential limitations of current models of cardiac G&R that need to be addressed before they can be utilized in clinical care. Finally, we briefly discuss the next feasible steps and future directions that could advance the field of cardiac G&R.
Collapse
Affiliation(s)
- Hossein Sharifi
- Department of Mechanical Engineering, University of Kentucky, 269 Ralph G. Anderson Building, Lexington, KY 40506-0503 USA
| | - Charles K. Mann
- Department of Mechanical Engineering, University of Kentucky, 269 Ralph G. Anderson Building, Lexington, KY 40506-0503 USA
| | - Alexus L. Rockward
- Department of Mechanical Engineering, University of Kentucky, 269 Ralph G. Anderson Building, Lexington, KY 40506-0503 USA
| | - Mohammad Mehri
- Department of Mechanical Engineering, University of Kentucky, 269 Ralph G. Anderson Building, Lexington, KY 40506-0503 USA
| | - Joy Mojumder
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI USA
| | - Lik-Chuan Lee
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI USA
| | - Kenneth S. Campbell
- Department of Physiology & Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY USA
| | - Jonathan F. Wenk
- Department of Mechanical Engineering, University of Kentucky, 269 Ralph G. Anderson Building, Lexington, KY 40506-0503 USA
- Department of Surgery, University of Kentucky, Lexington, KY USA
| |
Collapse
|
39
|
Yu M, Lu JH, Le S, Yan J. Unexpected Low Mechanical Stability of Titin I27 Domain at Physiologically Relevant Temperature. J Phys Chem Lett 2021; 12:7914-7920. [PMID: 34384021 DOI: 10.1021/acs.jpclett.1c01309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The extensively studied immunoglobulin (Ig) domain I27 of the giant force-bearing protein titin has provided a basis for our current understanding of the structural stability, dynamics, and function of the numerous mechanically stretched Ig domains in the force-bearing I-band of titin. The current consensus is that titin I27 has a high mechanical stability characterized by very low unfolding rate (<10-3 s-1) in physiological force range and high unfolding forces (>100 pN) at typical physiological force loading rates from experiments at typical laboratory temperatures. Here, we report that when the temperature is increased from 23 to 37 °C, the unfolding rate of I27 drastically increases by ∼100-fold at the physiological level of forces, indicating a low mechanical stability of I27 at physiological conditions. The result provides new insights into the structural states and the associated functions of I27 and other similar titin I-band Ig domains.
Collapse
Affiliation(s)
- Miao Yu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Jung-Hsuan Lu
- Department of Physics, National University of Singapore, Singapore 117542, Singapore
| | - Shimin Le
- Department of Physics, National University of Singapore, Singapore 117542, Singapore
| | - Jie Yan
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Department of Physics, National University of Singapore, Singapore 117542, Singapore
| |
Collapse
|
40
|
Alegre-Cebollada J. Protein nanomechanics in biological context. Biophys Rev 2021; 13:435-454. [PMID: 34466164 PMCID: PMC8355295 DOI: 10.1007/s12551-021-00822-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
How proteins respond to pulling forces, or protein nanomechanics, is a key contributor to the form and function of biological systems. Indeed, the conventional view that proteins are able to diffuse in solution does not apply to the many polypeptides that are anchored to rigid supramolecular structures. These tethered proteins typically have important mechanical roles that enable cells to generate, sense, and transduce mechanical forces. To fully comprehend the interplay between mechanical forces and biology, we must understand how protein nanomechanics emerge in living matter. This endeavor is definitely challenging and only recently has it started to appear tractable. Here, I introduce the main in vitro single-molecule biophysics methods that have been instrumental to investigate protein nanomechanics over the last 2 decades. Then, I present the contemporary view on how mechanical force shapes the free energy of tethered proteins, as well as the effect of biological factors such as post-translational modifications and mutations. To illustrate the contribution of protein nanomechanics to biological function, I review current knowledge on the mechanobiology of selected muscle and cell adhesion proteins including titin, talin, and bacterial pilins. Finally, I discuss emerging methods to modulate protein nanomechanics in living matter, for instance by inducing specific mechanical loss-of-function (mLOF). By interrogating biological systems in a causative manner, these new tools can contribute to further place protein nanomechanics in a biological context.
Collapse
|
41
|
Adewale AO, Ahn YH. Titin N2A Domain and Its Interactions at the Sarcomere. Int J Mol Sci 2021; 22:ijms22147563. [PMID: 34299183 PMCID: PMC8305307 DOI: 10.3390/ijms22147563] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/16/2022] Open
Abstract
Titin is a giant protein in the sarcomere that plays an essential role in muscle contraction with actin and myosin filaments. However, its utility goes beyond mechanical functions, extending to versatile and complex roles in sarcomere organization and maintenance, passive force, mechanosensing, and signaling. Titin’s multiple functions are in part attributed to its large size and modular structures that interact with a myriad of protein partners. Among titin’s domains, the N2A element is one of titin’s unique segments that contributes to titin’s functions in compliance, contraction, structural stability, and signaling via protein–protein interactions with actin filament, chaperones, stress-sensing proteins, and proteases. Considering the significance of N2A, this review highlights structural conformations of N2A, its predisposition for protein–protein interactions, and its multiple interacting protein partners that allow the modulation of titin’s biological effects. Lastly, the nature of N2A for interactions with chaperones and proteases is included, presenting it as an important node that impacts titin’s structural and functional integrity.
Collapse
|
42
|
Affiliation(s)
- Robbert J van der Pijl
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| | - Coen A C Ottenheijm
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| |
Collapse
|
43
|
van der Pijl RJ, van den Berg M, van de Locht M, Shen S, Bogaards SJP, Conijn S, Langlais P, Hooijman PE, Labeit S, Heunks LMA, Granzier H, Ottenheijm CAC. Muscle ankyrin repeat protein 1 (MARP1) locks titin to the sarcomeric thin filament and is a passive force regulator. J Gen Physiol 2021; 153:212403. [PMID: 34152365 PMCID: PMC8222902 DOI: 10.1085/jgp.202112925] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/19/2021] [Indexed: 12/12/2022] Open
Abstract
Muscle ankyrin repeat protein 1 (MARP1) is frequently up-regulated in stressed muscle, but its effect on skeletal muscle function is poorly understood. Here, we focused on its interaction with the titin–N2A element, found in titin’s molecular spring region. We show that MARP1 binds to F-actin, and that this interaction is stronger when MARP1 forms a complex with titin–N2A. Mechanics and super-resolution microscopy revealed that MARP1 “locks” titin–N2A to the sarcomeric thin filament, causing increased extension of titin’s elastic PEVK element and, importantly, increased passive force. In support of this mechanism, removal of thin filaments abolished the effect of MARP1 on passive force. The clinical relevance of this mechanism was established in diaphragm myofibers of mechanically ventilated rats and of critically ill patients. Thus, MARP1 regulates passive force by locking titin to the thin filament. We propose that in stressed muscle, this mechanism protects the sarcomere from mechanical damage.
Collapse
Affiliation(s)
- Robbert J van der Pijl
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tuscon, AZ
| | - Marloes van den Berg
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tuscon, AZ
| | - Martijn van de Locht
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Shengyi Shen
- Department of Cellular and Molecular Medicine, University of Arizona, Tuscon, AZ
| | - Sylvia J P Bogaards
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Stefan Conijn
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Paul Langlais
- Division of Endocrinology, University of Arizona, Tucson, AZ
| | - Pleuni E Hooijman
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Siegfried Labeit
- Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Leo M A Heunks
- Intensive Care Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tuscon, AZ
| | - Coen A C Ottenheijm
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tuscon, AZ
| |
Collapse
|
44
|
Thorolfsdottir RB, Sveinbjornsson G, Aegisdottir HM, Benonisdottir S, Stefansdottir L, Ivarsdottir EV, Halldorsson GH, Sigurdsson JK, Torp-Pedersen C, Weeke PE, Brunak S, Westergaard D, Pedersen OB, Sorensen E, Nielsen KR, Burgdorf KS, Banasik K, Brumpton B, Zhou W, Oddsson A, Tragante V, Hjorleifsson KE, Davidsson OB, Rajamani S, Jonsson S, Torfason B, Valgardsson AS, Thorgeirsson G, Frigge ML, Thorleifsson G, Norddahl GL, Helgadottir A, Gretarsdottir S, Sulem P, Jonsdottir I, Willer CJ, Hveem K, Bundgaard H, Ullum H, Arnar DO, Thorsteinsdottir U, Gudbjartsson DF, Holm H, Stefansson K. Genetic insight into sick sinus syndrome. Eur Heart J 2021; 42:1959-1971. [PMID: 36282123 PMCID: PMC8140484 DOI: 10.1093/eurheartj/ehaa1108] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/24/2020] [Accepted: 01/05/2021] [Indexed: 12/19/2022] Open
Abstract
Aims The aim of this study was to use human genetics to investigate the pathogenesis of sick sinus syndrome (SSS) and the role of risk factors in its development. Methods and results We performed a genome-wide association study of 6469 SSS cases and 1 000 187 controls from deCODE genetics, the Copenhagen Hospital Biobank, UK Biobank, and the HUNT study. Variants at six loci associated with SSS, a reported missense variant in MYH6, known atrial fibrillation (AF)/electrocardiogram variants at PITX2, ZFHX3, TTN/CCDC141, and SCN10A and a low-frequency (MAF = 1.1–1.8%) missense variant, p.Gly62Cys in KRT8 encoding the intermediate filament protein keratin 8. A full genotypic model best described the p.Gly62Cys association (P = 1.6 × 10−20), with an odds ratio (OR) of 1.44 for heterozygotes and a disproportionally large OR of 13.99 for homozygotes. All the SSS variants increased the risk of pacemaker implantation. Their association with AF varied and p.Gly62Cys was the only variant not associating with any other arrhythmia or cardiovascular disease. We tested 17 exposure phenotypes in polygenic score (PGS) and Mendelian randomization analyses. Only two associated with the risk of SSS in Mendelian randomization, AF, and lower heart rate, suggesting causality. Powerful PGS analyses provided convincing evidence against causal associations for body mass index, cholesterol, triglycerides, and type 2 diabetes (P > 0.05). Conclusion We report the associations of variants at six loci with SSS, including a missense variant in KRT8 that confers high risk in homozygotes and points to a mechanism specific to SSS development. Mendelian randomization supports a causal role for AF in the development of SSS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jon K Sigurdsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Christian Torp-Pedersen
- Department of Clinical Research and Cardiology, Nordsjaelland Hospital, Dyrehavevej 29, Hillerød 3400, Denmark
| | - Peter E Weeke
- Department of Cardiology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, Copenhagen 2200, Denmark
| | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, Copenhagen 2200, Denmark
| | - Ole B Pedersen
- Department of Clinical Immunology, Naestved Hospital, Ringstedgade 77B, Naestved 4700, Denmark
| | - Erik Sorensen
- Department of Clinical Immunology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Kaspar R Nielsen
- Department of Clinical Immunology, Aalborg University Hospital North, Urbansgade 36, Aalborg 9000, Denmark
| | - Kristoffer S Burgdorf
- Department of Clinical Immunology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, Copenhagen 2200, Denmark
| | - Ben Brumpton
- Department of Thoracic and Occupational Medicine, St. Olavs Hospital, Trondheim University Hospital, Prinsesse Kristinas gate 3, Trondheim 7030, Norway
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109-2218, USA
| | - Asmundur Oddsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | | | - Kristjan E Hjorleifsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Department of Computing and Mathematical Sciences, California Institute of Technology, 1200 E California Blvd. MC 305-16, Pasadena, CA 91125, USA
| | | | | | - Stefan Jonsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Bjarni Torfason
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Cardiothoracic Surgery, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Atli S Valgardsson
- Department of Cardiothoracic Surgery, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Gudmundur Thorgeirsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Medicine, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Michael L Frigge
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | | | | | - Anna Helgadottir
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | | | - Patrick Sulem
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Ingileif Jonsdottir
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Immunology, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Cristen J Willer
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109-2218, USA.,Department of Internal Medicine: Cardiology, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109 -5368, USA.,Department of Human Genetics, University of Michigan, 4909 Buhl Building, 1241 E. Catherine St., Ann Arbor, MI 48109 -5618, USA
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons gt. 1, Trondheim 7491, Norway.,Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postboks 8905, Trondheim 7491, Norway.,HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Forskningsveien 2, Levanger 7600, Norway
| | - Henning Bundgaard
- Department of Cardiology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Henrik Ullum
- Department of Clinical Immunology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark.,Statens Serum Institut, Artillerivej 5, Copenhagen 2300, Denmark
| | - David O Arnar
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Medicine, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Hjardarhagi 4, Reykjavik 107, Iceland
| | - Hilma Holm
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland
| | | |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW This review aims to give an update on recent findings related to the cardiac splicing factor RNA-binding motif protein 20 (RBM20) and RBM20 cardiomyopathy, a form of dilated cardiomyopathy caused by mutations in RBM20. RECENT FINDINGS While most research on RBM20 splicing targets has focused on titin (TTN), multiple studies over the last years have shown that other splicing targets of RBM20 including Ca2+/calmodulin-dependent kinase IIδ (CAMK2D) might be critically involved in the development of RBM20 cardiomyopathy. In this regard, loss of RBM20 causes an abnormal intracellular calcium handling, which may relate to the arrhythmogenic presentation of RBM20 cardiomyopathy. In addition, RBM20 presents clinically in a highly gender-specific manner, with male patients suffering from an earlier disease onset and a more severe disease progression. Further research on RBM20, and treatment of RBM20 cardiomyopathy, will need to consider both the multitude and relative contribution of the different splicing targets and related pathways, as well as gender differences.
Collapse
|
46
|
Zhou T, Fleming JR, Lange S, Hessel AL, Bogomolovas J, Stronczek C, Grundei D, Ghassemian M, Biju A, Börgeson E, Bullard B, Linke WA, Chen J, Kovermann M, Mayans O. Molecular Characterisation of Titin N2A and Its Binding of CARP Reveals a Titin/Actin Cross-linking Mechanism. J Mol Biol 2021; 433:166901. [PMID: 33647290 PMCID: PMC8052292 DOI: 10.1016/j.jmb.2021.166901] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/14/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022]
Abstract
Striated muscle responds to mechanical overload by rapidly up-regulating the expression of the cardiac ankyrin repeat protein, CARP, which then targets the sarcomere by binding to titin N2A in the I-band region. To date, the role of this interaction in the stress response of muscle remains poorly understood. Here, we characterise the molecular structure of the CARP-receptor site in titin (UN2A) and its binding of CARP. We find that titin UN2A contains a central three-helix bundle fold (ca 45 residues in length) that is joined to N- and C-terminal flanking immunoglobulin domains by long, flexible linkers with partial helical content. CARP binds titin by engaging an α-hairpin in the three-helix fold of UN2A, the C-terminal linker sequence, and the BC loop in Ig81, which jointly form a broad binding interface. Mutagenesis showed that the CARP/N2A association withstands sequence variations in titin N2A and we use this information to evaluate 85 human single nucleotide variants. In addition, actin co-sedimentation, co-transfection in C2C12 cells, proteomics on heart lysates, and the mechanical response of CARP-soaked myofibrils imply that CARP induces the cross-linking of titin and actin myofilaments, thereby increasing myofibril stiffness. We conclude that CARP acts as a regulator of force output in the sarcomere that preserves muscle mechanical performance upon overload stress.
Collapse
Affiliation(s)
- Tiankun Zhou
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | | | - Stephan Lange
- Division of Cardiology, School of Medicine, University of California, San Diego 92093, CA, USA; Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg 413 45, Sweden
| | - Anthony L Hessel
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Julius Bogomolovas
- School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Chiara Stronczek
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - David Grundei
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Majid Ghassemian
- Department of Chemistry and Biochemistry, University of California, San Diego 92093, CA, USA
| | - Andrea Biju
- Division of Cardiology, School of Medicine, University of California, San Diego 92093, CA, USA
| | - Emma Börgeson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg 413 45, Sweden
| | - Belinda Bullard
- Department of Biology, University of York, York YO10 5DD, UK
| | - Wolfgang A Linke
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Ju Chen
- School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael Kovermann
- Department of Chemistry, University of Konstanz, 78457 Konstanz, Germany.
| | - Olga Mayans
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany.
| |
Collapse
|
47
|
Stronczek C, Lange S, Bullard B, Wolniak S, Börgeson E, Mayans O, Fleming JR. The N2A region of titin has a unique structural configuration. J Gen Physiol 2021; 153:211969. [PMID: 33836065 PMCID: PMC8042602 DOI: 10.1085/jgp.202012766] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/09/2021] [Indexed: 01/04/2023] Open
Abstract
The N2A segment of titin is a main signaling hub in the sarcomeric I-band that recruits various signaling factors and processing enzymes. It has also been proposed to play a role in force production through its Ca2+-regulated association with actin. However, the molecular basis by which N2A performs these functions selectively within the repetitive and extensive titin chain remains poorly understood. Here, we analyze the structure of N2A components and their association with F-actin. Specifically, we characterized the structure of its Ig domains by elucidating the atomic structure of the I81-I83 tandem using x-ray crystallography and computing a homology model for I80. Structural data revealed these domains to present heterogeneous and divergent Ig folds, where I81 and I83 have unique loop structures. Notably, the I81-I83 tandem has a distinct rotational chain arrangement that confers it a unique multi-domain topography. However, we could not identify specific Ca2+-binding sites in these Ig domains, nor evidence of the association of titin N2A components with F-actin in transfected C2C12 myoblasts or C2C12-derived myotubes. In addition, F-actin cosedimentation assays failed to reveal binding to N2A. We conclude that N2A has a unique architecture that predictably supports its selective recruitment of binding partners in signaling, but that its mechanical role through interaction with F-actin awaits validation.
Collapse
Affiliation(s)
- Chiara Stronczek
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Stephan Lange
- Division of Cardiology, School of Medicine, University of California, San Diego, San Diego, CA.,Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Emma Börgeson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Olga Mayans
- Department of Biology, University of Konstanz, Konstanz, Germany
| | | |
Collapse
|
48
|
Solution NMR Structure of Titin N2A Region Ig Domain I83 and Its Interaction with Metal Ions. J Mol Biol 2021; 433:166977. [PMID: 33811919 DOI: 10.1016/j.jmb.2021.166977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/15/2021] [Accepted: 03/24/2021] [Indexed: 11/21/2022]
Abstract
Titin, the largest single chain protein known so far, has long been known to play a critical role in passive muscle function but recent studies have highlighted titin's role in active muscle function. One of the key elements in this role is the Ca2+-dependent interaction between titin's N2A region and the thin filament. An important element in this interaction is I83, the terminal immunoglobulin domain in the N2A region. There is limited structural information about this domain, but experimental evidence suggests that it plays a critical role in the N2A-actin binding interaction. We now report the solution NMR structure of I83 and characterize its dynamics and metal binding properties in detail. Its structure shows interesting relationships to other I-band Ig domains. Metal binding and dynamics data point towards the way the domain is evolutionarily optimized to interact with neighbouring domains. We also identify a calcium binding site on the N-terminal side of I83, which is expected to impact the interdomain interaction with the I82 domain. Together these results provide a first step towards a better understanding of the physiological effects associated with deletion of most of the I83 domain, as occurs in the mdm mouse model, as well as for future investigations of the N2A region.
Collapse
|
49
|
Mijailovich SM, Prodanovic M, Poggesi C, Geeves MA, Regnier M. Multiscale modeling of twitch contractions in cardiac trabeculae. J Gen Physiol 2021; 153:e202012604. [PMID: 33512405 PMCID: PMC7852458 DOI: 10.1085/jgp.202012604] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 08/31/2020] [Accepted: 11/20/2020] [Indexed: 12/23/2022] Open
Abstract
Understanding the dynamics of a cardiac muscle twitch contraction is complex because it requires a detailed understanding of the kinetic processes of the Ca2+ transient, thin-filament activation, and the myosin-actin cross-bridge chemomechanical cycle. Each of these steps has been well defined individually, but understanding how all three of the processes operate in combination is a far more complex problem. Computational modeling has the potential to provide detailed insight into each of these processes, how the dynamics of each process affect the complexity of contractile behavior, and how perturbations such as mutations in sarcomere proteins affect the complex interactions of all of these processes. The mechanisms involved in relaxation of tension during a cardiac twitch have been particularly difficult to discern due to nonhomogeneous sarcomere lengthening during relaxation. Here we use the multiscale MUSICO platform to model rat trabecular twitches. Validation of computational models is dependent on being able to simulate different experimental datasets, but there has been a paucity of data that can provide all of the required parameters in a single experiment, such as simultaneous measurements of force, intracellular Ca2+ transients, and sarcomere length dynamics. In this study, we used data from different studies collected under similar experimental conditions to provide information for all the required parameters. Our simulations established that twitches either in an isometric sarcomere or in fixed-length, multiple-sarcomere trabeculae replicate the experimental observations if models incorporate a length-tension relationship for the nonlinear series elasticity of muscle preparations and a scheme for thick-filament regulation. The thick-filament regulation assumes an off state in which myosin heads are parked onto the thick-filament backbone and are unable to interact with actin, a state analogous to the super-relaxed state. Including these two mechanisms provided simulations that accurately predict twitch contractions over a range of different conditions.
Collapse
Affiliation(s)
| | - Momcilo Prodanovic
- Bioengineering Research and Development Center, Kragujevac, Serbia
- Faculty of Engineering, University of Kragujevac, Kragujevac, Serbia
| | - Corrado Poggesi
- Department of Experimental & Clinical Medicine, University of Florence, Florence, Italy
| | | | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA
| |
Collapse
|
50
|
Münch J, Abdelilah-Seyfried S. Sensing and Responding of Cardiomyocytes to Changes of Tissue Stiffness in the Diseased Heart. Front Cell Dev Biol 2021; 9:642840. [PMID: 33718383 PMCID: PMC7952448 DOI: 10.3389/fcell.2021.642840] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/09/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiomyocytes are permanently exposed to mechanical stimulation due to cardiac contractility. Passive myocardial stiffness is a crucial factor, which defines the physiological ventricular compliance and volume of diastolic filling with blood. Heart diseases often present with increased myocardial stiffness, for instance when fibrotic changes modify the composition of the cardiac extracellular matrix (ECM). Consequently, the ventricle loses its compliance, and the diastolic blood volume is reduced. Recent advances in the field of cardiac mechanobiology revealed that disease-related environmental stiffness changes cause severe alterations in cardiomyocyte cellular behavior and function. Here, we review the molecular mechanotransduction pathways that enable cardiomyocytes to sense stiffness changes and translate those into an altered gene expression. We will also summarize current knowledge about when myocardial stiffness increases in the diseased heart. Sophisticated in vitro studies revealed functional changes, when cardiomyocytes faced a stiffer matrix. Finally, we will highlight recent studies that described modulations of cardiac stiffness and thus myocardial performance in vivo. Mechanobiology research is just at the cusp of systematic investigations related to mechanical changes in the diseased heart but what is known already makes way for new therapeutic approaches in regenerative biology.
Collapse
Affiliation(s)
- Juliane Münch
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.,Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| |
Collapse
|