1
|
Wang Y, Liu C, Fan Y, Niu C, Huang W, Pan Y, Li J, Wang Y, Li J. A multi-modal deep learning solution for precise pneumonia diagnosis: the PneumoFusion-Net model. Front Physiol 2025; 16:1512835. [PMID: 40144549 PMCID: PMC11937601 DOI: 10.3389/fphys.2025.1512835] [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: 10/17/2024] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
Background Pneumonia is considered one of the most important causes of morbidity and mortality in the world. Bacterial and viral pneumonia share many similar clinical features, thus making diagnosis a challenging task. Traditional diagnostic method developments mainly rely on radiological imaging and require a certain degree of consulting clinical experience, which can be inefficient and inconsistent. Deep learning for the classification of pneumonia in multiple modalities, especially integrating multiple data, has not been well explored. Methods The study introduce the PneumoFusion-Net, a deep learning-based multimodal framework that incorporates CT images, clinical text, numerical lab test results, and radiology reports for improved diagnosis. In the experiments, a dataset of 10,095 pneumonia CT images was used-including associated clinical data-most of which was used for training and validation while keeping part of it for validation on a held-out test set. Five-fold cross-validation was considered in order to evaluate this model, calculating different metrics including accuracy and F1-Score. Results PneumoFusion-Net, which achieved 98.96% classification accuracy with a 98% F1-score on the held-out test set, is highly effective in distinguishing bacterial from viral types of pneumonia. This has been highly beneficial for diagnosis, reducing misdiagnosis and further improving homogeneity across various data sets from multiple patients. Conclusion PneumoFusion-Net offers an effective and efficient approach to pneumonia classification by integrating diverse data sources, resulting in high diagnostic accuracy. Its potential for clinical integration could significantly reduce the burden of pneumonia diagnosis by providing radiologists and clinicians with a robust, automated diagnostic tool.
Collapse
Affiliation(s)
- Yujie Wang
- College of Information Engineering, Sichuan Agricultural University, Ya’an, China
- Deep Vision Agriculture Lab, Sichuan Agricultural University, Ya’an, China
| | - Can Liu
- College of Information Engineering, Sichuan Agricultural University, Ya’an, China
| | - Yinghan Fan
- College of Information Engineering, Sichuan Agricultural University, Ya’an, China
| | - Chenyue Niu
- College of Information Engineering, Sichuan Agricultural University, Ya’an, China
| | - Wanyun Huang
- College of Information Engineering, Sichuan Agricultural University, Ya’an, China
| | - Yixuan Pan
- College of Science, Sichuan Agricultural University, Ya’an, China
| | - Jingze Li
- College of Information Engineering, Sichuan Agricultural University, Ya’an, China
- Deep Vision Agriculture Lab, Sichuan Agricultural University, Ya’an, China
| | - Yilin Wang
- College of Information Engineering, Sichuan Agricultural University, Ya’an, China
| | - Jun Li
- College of Information Engineering, Sichuan Agricultural University, Ya’an, China
- Agricultural Information Engineering Higher Institution Key Laboratory of Sichuan Province, Ya’an, China
- Ya’an Digital Agricultural Engineering Technology Research Center, Ya’an, China
| |
Collapse
|
2
|
Støle TP, Lunde M, Gehmlich K, Christensen G, Louch WE, Carlson CR. Exploring Syndecan-4 and MLP and Their Interaction in Primary Cardiomyocytes and H9c2 Cells. Cells 2024; 13:947. [PMID: 38891079 PMCID: PMC11172336 DOI: 10.3390/cells13110947] [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: 03/25/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
The transmembrane proteoglycan syndecan-4 is known to be involved in the hypertrophic response to pressure overload. Although multiple downstream signaling pathways have been found to be involved in this response in a syndecan-4-dependent manner, there are likely more signaling components involved. As part of a larger syndecan-4 interactome screening, we have previously identified MLP as a binding partner to the cytoplasmic tail of syndecan-4. Interestingly, many human MLP mutations have been found in patients with hypertrophic (HCM) and dilated cardiomyopathy (DCM). To gain deeper insight into the role of the syndecan-4-MLP interaction and its potential involvement in MLP-associated cardiomyopathy, we have here investigated the syndecan-4-MLP interaction in primary adult rat cardiomyocytes and the H9c2 cell line. The binding of syndecan-4 and MLP was analyzed in total lysates and subcellular fractions of primary adult rat cardiomyocytes, and baseline and differentiated H9c2 cells by immunoprecipitation. MLP and syndecan-4 localization were determined by confocal microscopy, and MLP oligomerization was determined by immunoblotting under native conditions. Syndecan-4-MLP binding, as well as MLP self-association, were also analyzed by ELISA and peptide arrays. Our results showed that MLP-WT and syndecan-4 co-localized in many subcellular compartments; however, their binding was only detected in nuclear-enriched fractions of isolated adult cardiomyocytes. In vitro, syndecan-4 bound to MLP at three sites, and this binding was reduced in some HCM-associated MLP mutations. While MLP and syndecan-4 also co-localized in many subcellular fractions of H9c2 cells, these proteins did not bind at baseline or after differentiation into cardiomyocyte-resembling cells. Independently of syndecan-4, mutated MLP proteins had an altered subcellular localization in H9c2 cells, compared to MLP-WT. The DCM- and HCM-associated MLP mutations, W4R, L44P, C58G, R64C, Y66C, K69R, G72R, and Q91L, affected the oligomerization of MLP with an increase in monomeric at the expense of trimeric and tetrameric recombinant MLP protein. Lastly, two crucial sites for MLP self-association were identified, which were reduced in most MLP mutations. Our data indicate that the syndecan-4-MLP interaction was present in nuclear-enriched fractions of isolated adult cardiomyocytes and that this interaction was disrupted by some HCM-associated MLP mutations. MLP mutations were also linked to changes in MLP oligomerization and self-association, which may be essential for its interaction with syndecan-4 and a critical molecular mechanism of MLP-associated cardiomyopathy.
Collapse
Affiliation(s)
- Thea Parsberg Støle
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; (M.L.); (G.C.); (W.E.L.); (C.R.C.)
| | - Marianne Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; (M.L.); (G.C.); (W.E.L.); (C.R.C.)
- K.G. Jebsen Center for Cardiac Research, University of Oslo, 0313 Oslo, Norway
| | - Katja Gehmlich
- Institute for Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford OX3 9DU, UK
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; (M.L.); (G.C.); (W.E.L.); (C.R.C.)
- K.G. Jebsen Center for Cardiac Research, University of Oslo, 0313 Oslo, Norway
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; (M.L.); (G.C.); (W.E.L.); (C.R.C.)
- K.G. Jebsen Center for Cardiac Research, University of Oslo, 0313 Oslo, Norway
| | - Cathrine Rein Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; (M.L.); (G.C.); (W.E.L.); (C.R.C.)
| |
Collapse
|
3
|
Sharif NA. Electrical, Electromagnetic, Ultrasound Wave Therapies, and Electronic Implants for Neuronal Rejuvenation, Neuroprotection, Axonal Regeneration, and IOP Reduction. J Ocul Pharmacol Ther 2023; 39:477-498. [PMID: 36126293 DOI: 10.1089/jop.2022.0046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The peripheral nervous system (PNS) of mammals and nervous systems of lower organisms possess significant regenerative potential. In contrast, although neural plasticity can provide some compensation, the central nervous system (CNS) neurons and nerves of adult mammals generally fail to regenerate after an injury or damage. However, use of diverse electrical, electromagnetic and sonographic energy waves are illuminating novel ways to stimulate neuronal differentiation, proliferation, neurite growth, and axonal elongation/regeneration leading to various levels of functional recovery in animals and humans afflicted with disorders of the CNS, PNS, retina, and optic nerve. Tools such as acupuncture, electroacupuncture, electroshock therapy, electrical stimulation, transcranial magnetic stimulation, red light therapy, and low-intensity pulsed ultrasound therapy are demonstrating efficacy in treating many different maladies. These include wound healing, partial recovery from motor dysfunctions, recovery from ischemic/reperfusion insults and CNS and ocular remyelination, retinal ganglion cell (RGC) rejuvenation, and RGC axonal regeneration. Neural rejuvenation and axonal growth/regeneration processes involve activation or intensifying of the intrinsic bioelectric waves (action potentials) that exist in every neuronal circuit of the body. In addition, reparative factors released at the nerve terminals and via neuronal dendrites (transmitter substances), extracellular vesicles containing microRNAs and neurotrophins, and intercellular communication occurring via nanotubes aid in reestablishing lost or damaged connections between the traumatized tissues and the PNS and CNS. Many other beneficial effects of the aforementioned treatment paradigms are mediated via gene expression alterations such as downregulation of inflammatory and death-signal genes and upregulation of neuroprotective and cytoprotective genes. These varied techniques and technologies will be described and discussed covering cell-based and animal model-based studies. Data from clinical applications and linkage to human ocular diseases will also be discussed where relevant translational research has been reported.
Collapse
Affiliation(s)
- Najam A Sharif
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, California, USA
- Singapore Eye Research Institute (SERI), Singapore
- SingHealth Duke-NUS Ophthalmology and Visual Sciences Academic Clinical Programme, Duke-National University of Singapore Medical School, Singapore
- Department of Surgery and Cancer, Imperial College of Science and Technology, London, United Kingdom
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center, Fort Worth, Texas, USA
- Department of Pharmacy Sciences, Creighton University, Omaha, Nebraska, USA
- Insitute of Ophthalmology, University College London (UCL), London, United Kingdom
| |
Collapse
|
4
|
Holmes JB, Lemieux ME, Stelzer JE. Torsional and strain dysfunction precede overt heart failure in a mouse model of dilated cardiomyopathy pathogenesis. Am J Physiol Heart Circ Physiol 2023; 325:H449-H467. [PMID: 37417875 PMCID: PMC10538988 DOI: 10.1152/ajpheart.00130.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/24/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023]
Abstract
Detailed assessments of whole heart mechanics are crucial for understanding the consequences of sarcomere perturbations that lead to cardiomyopathy in mice. Echocardiography offers an accessible and cost-effective method of obtaining metrics of cardiac function, but the most routine imaging and analysis protocols might not identify subtle mechanical deficiencies. This study aims to use advanced echocardiography imaging and analysis techniques to identify previously unappreciated mechanical deficiencies in a mouse model of dilated cardiomyopathy (DCM) before the onset of overt systolic heart failure (HF). Mice lacking muscle LIM protein expression (MLP-/-) were used to model DCM-linked HF pathogenesis. Left ventricular (LV) function of MLP-/- and wild-type (WT) controls were studied at 3, 6, and 10 wk of age using conventional and four-dimensional (4-D) echocardiography, followed by speckle-tracking analysis to assess torsional and strain mechanics. Mice were also studied with RNA-seq. Although 3-wk-old MLP-/- mice showed normal LV ejection fraction (LVEF), these mice displayed abnormal torsional and strain mechanics alongside reduced β-adrenergic reserve. Transcriptome analysis showed that these defects preceded most molecular markers of HF. However, these markers became upregulated as MLP-/- mice aged and developed overt systolic dysfunction. These findings indicate that subtle deficiencies in LV mechanics, undetected by LVEF and conventional molecular markers, may act as pathogenic stimuli in DCM-linked HF. Using these analyses in future studies will further help connect in vitro measurements of the sarcomere function to whole heart function.NEW & NOTEWORTHY A detailed study of how perturbations to sarcomere proteins impact whole heart mechanics in mouse models is a major yet challenging step in furthering our understanding of cardiovascular pathophysiology. This study uses advanced echocardiographic imaging and analysis techniques to reveal previously unappreciated subclinical whole heart mechanical defects in a mouse model of cardiomyopathy. In doing so, it offers an accessible set of measurements for future studies to use when connecting sarcomere and whole heart function.
Collapse
Affiliation(s)
- Joshua B Holmes
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, United States
| | | | - Julian E Stelzer
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
5
|
Tian S, Guo L, Song Y, Miao J, Peng M, Fang X, Bai M, Miao M. Transcriptomic analysis the mechanisms of anti-osteoporosis of desert-living Cistanche herb in ovariectomized rats of postmenopausal osteoporosis. Funct Integr Genomics 2023; 23:237. [PMID: 37439895 DOI: 10.1007/s10142-023-01154-5] [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: 04/08/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/14/2023]
Abstract
Desert-living Cistanche herb (DC), as a traditional Chinese medicine for tonifying kidney yang, is often used to treat postmenopausal osteoporosis (PMOP). Total phenylethanoid glycosides are instruction ingredients for discrimination and assay according to the China pharmacopoeia for DC. This research aimed to reveal the anti-osteoporosis mechanism of total phenylethanoid glycosides of DC (PGC) by transcriptomic analysis of ovariectomized rats. Serum levels of BGP were evaluated by ELISA, the bone weight was measured, and transmission electron microscopy was used to examine the ultrastructure of osteoblasts in rats. In addition, micro-CT was used to detect the bone volume (Tb.BS/BV), bone mineral density (Tb.BMD), and bone mineral content (Tb.BMC) in trabecular bone, and the ratio of cortical bone area to total area (Ct.ar/Tt.ar), and the level of bone mineral content (Ct.BMC) in cortical bone. Differential expressed genes (DEGs) after PGC treatment were analyzed by transcriptomics. Then, a bioinformatics analysis of DEGs was carried out through GO enrichment, KEGG enrichment, and selection of the nucleus gene through the protein-protein interaction network. Through qRT-PCR analysis, the DEGs were verified. The analysis results indicated that PGC increased the secretion of osteogenic markers, and ultrastructural characterization of osteoblasts and bone morphology were improved in ovariectomized rats. A total of 269 genes were differentially expressed, including 201 genes that were downregulated and 68 genes that were upregulated between the model group and the PGC group. Bioinformation analysis results prompt the conclusion that PGC could promote the bone metabolism by muscle cell development, myofibril assembly, etc. In addition, our study also found that PGC has a good effect on osteoporosis complicated with cardiomyopathy, and it also provided evidence for the correlation between sarcopenia and osteoporosis.
Collapse
Affiliation(s)
- Shuo Tian
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Henan Collaborative Innovation Center for Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zheng Zhou, 450046, China
| | - Lin Guo
- Department of Pharmacology, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yagang Song
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jinxin Miao
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Mengfan Peng
- Department of Pharmacology, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xiaoyan Fang
- Department of Pharmacology, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Ming Bai
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Mingsan Miao
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
6
|
Chauhan PK, Sowdhamini R. Computational analysis of the flexibility in the disordered linker region connecting LIM domains in cysteine–glycine-rich protein. Front Genet 2023; 14:1134509. [PMID: 37065494 PMCID: PMC10090389 DOI: 10.3389/fgene.2023.1134509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/06/2023] [Indexed: 03/31/2023] Open
Abstract
One of the key proteins that are present in the Z-disc of cardiac tissues, CSRP3, has been implicated in dilated and hypertrophic cardiomyopathy leading to heart failure. Although multiple cardiomyopathy-related mutations have been reported to reside on the two LIM domains and the disordered regions connecting the domains in this protein, the exact role of the disordered linker region is not clear. The linker harbors a few post-translational modification sites and is expected to be a regulatory site. We have carried out evolutionary studies on 5614 homologs spanning across taxa. We also performed molecular dynamics simulations of full-length CSRP3 to show that the length variations and conformational flexibility of the disordered linker could provide additional levels of functional modulation. Finally, we show that the CSRP3 homologs with widely different lengths of the linker regions could display diversity in their functional specifications. The present study provides a useful perspective to our understanding of the evolution of the disordered region between CSRP3 LIM domains.
Collapse
Affiliation(s)
- Pankaj Kumar Chauhan
- National Centre for Biological Sciences Tata Institute of Fundamental Research, Bangalore Karnataka, India
| | - R. Sowdhamini
- National Centre for Biological Sciences Tata Institute of Fundamental Research, Bangalore Karnataka, India
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
- *Correspondence: R. Sowdhamini,
| |
Collapse
|
7
|
Chapman EA, Aballo TJ, Melby JA, Zhou T, Price SJ, Rossler KJ, Lei I, Tang PC, Ge Y. Defining the Sarcomeric Proteoform Landscape in Ischemic Cardiomyopathy by Top-Down Proteomics. J Proteome Res 2023; 22:931-941. [PMID: 36800490 PMCID: PMC10115148 DOI: 10.1021/acs.jproteome.2c00729] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Ischemic cardiomyopathy (ICM) is a prominent form of heart failure, but the molecular mechanisms underlying ICM remain relatively understudied due to marked phenotypic heterogeneity. Alterations in post-translational modifications (PTMs) and isoform switches in sarcomeric proteins play important roles in cardiac pathophysiology. Thus, it is essential to define sarcomeric proteoform landscape to better understand ICM. Herein, we have implemented a top-down liquid chromatography (LC)-mass spectrometry (MS)-based proteomics method for the identification and quantification of sarcomeric proteoforms in the myocardia of donors without heart diseases (n = 16) compared to end-stage ICM patients (n = 16). Importantly, quantification of post-translational modifications (PTMs) and expression reveal significant changes in various sarcomeric proteins extracted from ICM tissues. Changes include altered phosphorylation and expression of cardiac troponin I (cTnI) and enigma homologue 2 (ENH2) as well as an increase in muscle LIM protein (MLP) and calsarcin-1 (Cal-1) phosphorylation in ICM hearts. Our results imply that the contractile apparatus of the sarcomere is severely dysregulated during ICM. Thus, this is the first study to uncover significant molecular changes to multiple sarcomeric proteins in the LV myocardia of the end-stage ICM patients using liquid chromatography-mass spectrometry (LC-MS)-based top-down proteomics. Raw data are available via the PRIDE repository with identifier PXD038066.
Collapse
Affiliation(s)
- Emily A. Chapman
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Timothy J. Aballo
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Jake A. Melby
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Tianhua Zhou
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Scott J. Price
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Kalina J. Rossler
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Ienglam Lei
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Paul C. Tang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| |
Collapse
|
8
|
Noureddine M, Gehmlich K. Structural and signaling proteins in the Z-disk and their role in cardiomyopathies. Front Physiol 2023; 14:1143858. [PMID: 36935760 PMCID: PMC10017460 DOI: 10.3389/fphys.2023.1143858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The sarcomere is the smallest functional unit of muscle contraction. It is delineated by a protein-rich structure known as the Z-disk, alternating with M-bands. The Z-disk anchors the actin-rich thin filaments and plays a crucial role in maintaining the mechanical stability of the cardiac muscle. A multitude of proteins interact with each other at the Z-disk and they regulate the mechanical properties of the thin filaments. Over the past 2 decades, the role of the Z-disk in cardiac muscle contraction has been assessed widely, however, the impact of genetic variants in Z-disk proteins has still not been fully elucidated. This review discusses the various Z-disk proteins (alpha-actinin, filamin C, titin, muscle LIM protein, telethonin, myopalladin, nebulette, and nexilin) and Z-disk-associated proteins (desmin, and obscurin) and their role in cardiac structural stability and intracellular signaling. This review further explores how genetic variants of Z-disk proteins are linked to inherited cardiac conditions termed cardiomyopathies.
Collapse
Affiliation(s)
- Maya Noureddine
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
9
|
McNamara JW, Parker BL, Voges HK, Mehdiabadi NR, Bolk F, Ahmad F, Chung JD, Charitakis N, Molendijk J, Zech ATL, Lal S, Ramialison M, Karavendzas K, Pointer HL, Syrris P, Lopes LR, Elliott PM, Lynch GS, Mills RJ, Hudson JE, Watt KI, Porrello ER, Elliott DA. Alpha kinase 3 signaling at the M-band maintains sarcomere integrity and proteostasis in striated muscle. NATURE CARDIOVASCULAR RESEARCH 2023; 2:159-173. [PMID: 39196058 PMCID: PMC11358020 DOI: 10.1038/s44161-023-00219-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/19/2023] [Indexed: 08/29/2024]
Abstract
Muscle contraction is driven by the molecular machinery of the sarcomere. As phosphorylation is a critical regulator of muscle function, the identification of regulatory kinases is important for understanding sarcomere biology. Pathogenic variants in alpha kinase 3 (ALPK3) cause cardiomyopathy and musculoskeletal disease, but little is known about this atypical kinase. Here we show that ALPK3 is an essential component of the M-band of the sarcomere and define the ALPK3-dependent phosphoproteome. ALPK3 deficiency impaired contractility both in human cardiac organoids and in the hearts of mice harboring a pathogenic truncating Alpk3 variant. ALPK3-dependent phosphopeptides were enriched for sarcomeric components of the M-band and the ubiquitin-binding protein sequestosome-1 (SQSTM1) (also known as p62). Analysis of the ALPK3 interactome confirmed binding to M-band proteins including SQSTM1. In human pluripotent stem cell-derived cardiomyocytes modeling cardiomyopathic ALPK3 mutations, sarcomeric organization and M-band localization of SQSTM1 were abnormal suggesting that this mechanism may underly disease pathogenesis.
Collapse
Affiliation(s)
- James W McNamara
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Benjamin L Parker
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Holly K Voges
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Neda R Mehdiabadi
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Francesca Bolk
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Feroz Ahmad
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Jin D Chung
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Natalie Charitakis
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Jeffrey Molendijk
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Antonia T L Zech
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Sean Lal
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia
| | - Mirana Ramialison
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Kathy Karavendzas
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Hayley L Pointer
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Petros Syrris
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, UK
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Luis R Lopes
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, UK
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Perry M Elliott
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, UK
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Gordon S Lynch
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Richard J Mills
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - James E Hudson
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kevin I Watt
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Enzo R Porrello
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia.
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia.
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
| | - David A Elliott
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
10
|
Yamada S, Ko T, Hatsuse S, Nomura S, Zhang B, Dai Z, Inoue S, Kubota M, Sawami K, Yamada T, Sassa T, Katagiri M, Fujita K, Katoh M, Ito M, Harada M, Toko H, Takeda N, Morita H, Aburatani H, Komuro I. Spatiotemporal transcriptome analysis reveals critical roles for mechano-sensing genes at the border zone in remodeling after myocardial infarction. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1072-1083. [PMID: 39195917 PMCID: PMC11358009 DOI: 10.1038/s44161-022-00140-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/02/2022] [Indexed: 08/29/2024]
Abstract
The underlying mechanisms of ventricular remodeling after myocardial infarction (MI) remain largely unknown. In this study, we performed an integrative analysis of spatial transcriptomics and single-nucleus RNA sequencing (snRNA-seq) in a murine MI model and found that mechanical stress-response genes are expressed at the border zone and play a critical role in left ventricular remodeling after MI. An integrative analysis of snRNA-seq and spatial transcriptome of the heart tissue after MI identified the unique cluster that appeared at the border zone in an early stage, highly expressing mechano-sensing genes, such as Csrp3. AAV9-mediated gene silencing and overexpression of Csrp3 demonstrated that upregulation of Csrp3 plays critical roles in preventing cardiac remodeling after MI by regulation of genes associated with mechano-sensing. Overall, our study not only provides an insight into spatiotemporal molecular changes after MI but also highlights that the mechano-sensing genes at the border zone act as adaptive regulators of left ventricular remodeling.
Collapse
Affiliation(s)
- Shintaro Yamada
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan
| | - Toshiyuki Ko
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Satoshi Hatsuse
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan.
| | - Bo Zhang
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan
| | - Zhehao Dai
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan
| | - Shunsuke Inoue
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan
| | - Masayuki Kubota
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kosuke Sawami
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan
| | - Takanobu Yamada
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan
| | - Tatsuro Sassa
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan
| | - Mikako Katagiri
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kanna Fujita
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan
| | - Manami Katoh
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan
| | - Masamichi Ito
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Mutsuo Harada
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Haruhiro Toko
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Norifumi Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technologies, University of Tokyo, Tokyo, Japan.
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
11
|
Germain P, Delalande A, Pichon C. Role of Muscle LIM Protein in Mechanotransduction Process. Int J Mol Sci 2022; 23:ijms23179785. [PMID: 36077180 PMCID: PMC9456170 DOI: 10.3390/ijms23179785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/14/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022] Open
Abstract
The induction of protein synthesis is crucial to counteract the deconditioning of neuromuscular system and its atrophy. In the past, hormones and cytokines acting as growth factors involved in the intracellular events of these processes have been identified, while the implications of signaling pathways associated with the anabolism/catabolism ratio in reference to the molecular mechanism of skeletal muscle hypertrophy have been recently identified. Among them, the mechanotransduction resulting from a mechanical stress applied to the cell appears increasingly interesting as a potential pathway for therapeutic intervention. At present, there is an open question regarding the type of stress to apply in order to induce anabolic events or the type of mechanical strain with respect to the possible mechanosensing and mechanotransduction processes involved in muscle cells protein synthesis. This review is focused on the muscle LIM protein (MLP), a structural and mechanosensing protein with a LIM domain, which is expressed in the sarcomere and costamere of striated muscle cells. It acts as a transcriptional cofactor during cell proliferation after its nuclear translocation during the anabolic process of differentiation and rebuilding. Moreover, we discuss the possible opportunity of stimulating this mechanotransduction process to counteract the muscle atrophy induced by anabolic versus catabolic disorders coming from the environment, aging or myopathies.
Collapse
Affiliation(s)
- Philippe Germain
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
| | - Anthony Delalande
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
| | - Chantal Pichon
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
- Institut Universitaire de France, 1 Rue Descartes, 75231 Paris, France
- Correspondence:
| |
Collapse
|
12
|
Zhuang C, Chen R, Zheng Z, Lu J, Hong C. Toll-Like Receptor 3 in Cardiovascular Diseases. Heart Lung Circ 2022; 31:e93-e109. [PMID: 35367134 DOI: 10.1016/j.hlc.2022.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023]
Abstract
Toll-like receptor 3 (TLR3) is an important member of the innate immune response receptor toll-like receptors (TLRs) family, which plays a vital role in regulating immune response, promoting the maturation and differentiation of immune cells, and participating in the response of pro-inflammatory factors. TLR3 is activated by pathogen-associated molecular patterns and damage-associated molecular patterns, which support the pathophysiology of many diseases related to inflammation. An increasing number of studies have confirmed that TLR3, as a crucial medium of innate immunity, participates in the occurrence and development of cardiovascular diseases (CVDs) by regulating the transcription and translation of various cytokines, thus affecting the structure and physiological function of resident cells in the cardiovascular system, including vascular endothelial cells, vascular smooth muscle cells, cardiomyocytes, fibroblasts and macrophages. The dysfunction and structural damage of vascular endothelial cells and proliferation of vascular smooth muscle cells are the key factors in the occurrence of vascular diseases such as pulmonary arterial hypertension, atherosclerosis, myocardial hypertrophy, myocardial infarction, ischaemia/reperfusion injury, and heart failure. Meanwhile, cardiomyocytes, fibroblasts, and macrophages are involved in the development of CVDs. Therefore, the purpose of this review was to explore the latest research published on TLR3 in CVDs and discuss current understanding of potential mechanisms by which TLR3 contributes to CVDs. Even though TLR3 is a developing area, it has strong treatment potential as an immunomodulator and deserves further study for clinical translation.
Collapse
Affiliation(s)
- Chunying Zhuang
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; First Clinical School, Guangzhou Medical University, Guangzhou, China
| | - Riken Chen
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenzhen Zheng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Guangzhou, China
| | - Jianmin Lu
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cheng Hong
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
13
|
The focal adhesion protein β-parvin controls cardiomyocyte shape and sarcomere assembly in response to mechanical load. Curr Biol 2022; 32:3033-3047.e9. [PMID: 35688156 DOI: 10.1016/j.cub.2022.05.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/12/2022] [Accepted: 05/17/2022] [Indexed: 01/01/2023]
Abstract
Physiological and pathological cardiac stress induced by exercise and hypertension, respectively, increase the hemodynamic load for the heart and trigger specific hypertrophic signals in cardiomyocytes leading to adaptive or maladaptive cardiac hypertrophy responses involving a mechanosensitive remodeling of the contractile cytoskeleton. Integrins sense load and have been implicated in cardiac hypertrophy, but how they discriminate between the two types of cardiac stress and translate mechanical loads into specific cytoskeletal signaling pathways is not clear. Here, we report that the focal adhesion protein β-parvin is highly expressed in cardiomyocytes and facilitates the formation of cell protrusions, the serial assembly of newly synthesized sarcomeres, and the hypertrophic growth of neonatal rat ventricular cardiomyocytes (NRVCs) in vitro. In addition, physiological mechanical loading of NRVCs by either the application of cyclic, uni-axial stretch, or culture on physiologically stiff substrates promotes NRVC elongation in a β-parvin-dependent manner, which is achieved by binding of β-parvin to α/β-PIX, which in turn activates Rac1. Importantly, loss-of-function studies in mice also revealed that β-parvin is essential for the exercise-induced cardiac hypertrophy response in vivo. Our results identify β-parvin as a novel mechano-responsive signaling hub in hypertrophic cardiomyocytes that drives cell elongation in response to physiological mechanical loads.
Collapse
|
14
|
Impact of Treadmill Interval Running on the Appearance of Zinc Finger Protein FHL2 in Bone Marrow Cells in a Rat Model: A Pilot Study. Life (Basel) 2022; 12:life12040528. [PMID: 35455019 PMCID: PMC9029125 DOI: 10.3390/life12040528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Although the benefits of physical exercise to preserve bone quality are now widely recognized, the intimate mechanisms leading to the underlying cell responses still require further investigations. Interval training running, for instance, appears as a generator of impacts on the skeleton, and particularly on the progenitor cells located in the bone marrow. Therefore, if this kind of stimulus initiates bone cell proliferation and differentiation, the activation of a devoted signaling pathway by mechano-transduction seems likely. This study aimed at investigating the effects of an interval running program on the appearance of the zinc finger protein FHL2 in bone cells and their anatomical location. Twelve 5-week-old male Wistar rats were randomly allocated to one of the following groups (n = 6 per group): sedentary control (SED) or high-intensity interval running (EX, 8 consecutive weeks). FHL2 identification in bone cells was performed by immuno-histochemistry on serial sections of radii. We hypothesized that impacts generated by running could activate, in vivo, a specific signaling pathway, through an integrin-mediated mechano-transductive process, leading to the synthesis of FHL2 in bone marrow cells. Our data demonstrated the systematic appearance of FHL2 (% labeled cells: 7.5%, p < 0.001) in bone marrow obtained from EX rats, whereas no FHL2 was revealed in SED rats. These results suggest that the mechanical impacts generated during high-intensity interval running activate a signaling pathway involving nuclear FHL2, such as that also observed with dexamethasone administration. Consequently, interval running could be proposed as a non-pharmacological strategy to contribute to bone marrow cell osteogenic differentiation.
Collapse
|
15
|
Solís C, Russell B. Striated muscle proteins are regulated both by mechanical deformation and by chemical post-translational modification. Biophys Rev 2021; 13:679-695. [PMID: 34777614 PMCID: PMC8555064 DOI: 10.1007/s12551-021-00835-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/23/2021] [Indexed: 01/09/2023] Open
Abstract
All cells sense force and build their cytoskeleton to optimize function. How is this achieved? Two major systems are involved. The first is that load deforms specific protein structures in a proportional and orientation-dependent manner. The second is post-translational modification of proteins as a consequence of signaling pathway activation. These two processes work together in a complex way so that local subcellular assembly as well as overall cell function are controlled. This review discusses many cell types but focuses on striated muscle. Detailed information is provided on how load deforms the structure of proteins in the focal adhesions and filaments, using α-actinin, vinculin, talin, focal adhesion kinase, LIM domain-containing proteins, filamin, myosin, titin, and telethonin as examples. Second messenger signals arising from external triggers are distributed throughout the cell causing post-translational or chemical modifications of protein structures, with the actin capping protein CapZ and troponin as examples. There are numerous unanswered questions of how mechanical and chemical signals are integrated by muscle proteins to regulate sarcomere structure and function yet to be studied. Therefore, more research is needed to see how external triggers are integrated with local tension generated within the cell. Nonetheless, maintenance of tension in the sarcomere is the essential and dominant mechanism, leading to the well-known phrase in exercise physiology: "use it or lose it."
Collapse
Affiliation(s)
- Christopher Solís
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Brenda Russell
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| |
Collapse
|
16
|
Havlenova T, Skaroupkova P, Miklovic M, Behounek M, Chmel M, Jarkovska D, Sviglerova J, Stengl M, Kolar M, Novotny J, Benes J, Cervenka L, Petrak J, Melenovsky V. Right versus left ventricular remodeling in heart failure due to chronic volume overload. Sci Rep 2021; 11:17136. [PMID: 34429479 PMCID: PMC8384875 DOI: 10.1038/s41598-021-96618-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023] Open
Abstract
Mechanisms of right ventricular (RV) dysfunction in heart failure (HF) are poorly understood. RV response to volume overload (VO), a common contributing factor to HF, is rarely studied. The goal was to identify interventricular differences in response to chronic VO. Rats underwent aorto-caval fistula (ACF)/sham operation to induce VO. After 24 weeks, RV and left ventricular (LV) functions, gene expression and proteomics were studied. ACF led to biventricular dilatation, systolic dysfunction and hypertrophy affecting relatively more RV. Increased RV afterload contributed to larger RV stroke work increment compared to LV. Both ACF ventricles displayed upregulation of genes of myocardial stress and metabolism. Most proteins reacted to VO in a similar direction in both ventricles, yet the expression changes were more pronounced in RV (pslope: < 0.001). The most upregulated were extracellular matrix (POSTN, NRAP, TGM2, CKAP4), cell adhesion (NCAM, NRAP, XIRP2) and cytoskeletal proteins (FHL1, CSRP3) and enzymes of carbohydrate (PKM) or norepinephrine (MAOA) metabolism. Downregulated were MYH6 and FAO enzymes. Therefore, when exposed to identical VO, both ventricles display similar upregulation of stress and metabolic markers. Relatively larger response of ACF RV compared to the LV may be caused by concomitant pulmonary hypertension. No evidence supports RV chamber-specific regulation of protein expression in response to VO.
Collapse
Affiliation(s)
- Tereza Havlenova
- grid.418930.70000 0001 2299 1368Department of Cardiology, Institute for Clinical and Experimental Medicine - IKEM, Videnska 1958/9, 140 21 Prague 4, Czech Republic ,grid.4491.80000 0004 1937 116XDepartment of Pathophysiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petra Skaroupkova
- grid.418930.70000 0001 2299 1368Department of Cardiology, Institute for Clinical and Experimental Medicine - IKEM, Videnska 1958/9, 140 21 Prague 4, Czech Republic
| | - Matus Miklovic
- grid.418930.70000 0001 2299 1368Department of Cardiology, Institute for Clinical and Experimental Medicine - IKEM, Videnska 1958/9, 140 21 Prague 4, Czech Republic ,grid.4491.80000 0004 1937 116XDepartment of Pathophysiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Matej Behounek
- grid.4491.80000 0004 1937 116XBIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Chmel
- grid.4491.80000 0004 1937 116XBIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Dagmar Jarkovska
- grid.4491.80000 0004 1937 116XFaculty of Medicine in Pilsen, Charles University, Prague, Czech Republic
| | - Jitka Sviglerova
- grid.4491.80000 0004 1937 116XFaculty of Medicine in Pilsen, Charles University, Prague, Czech Republic
| | - Milan Stengl
- grid.4491.80000 0004 1937 116XFaculty of Medicine in Pilsen, Charles University, Prague, Czech Republic
| | - Michal Kolar
- grid.418827.00000 0004 0620 870XInstitute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Novotny
- grid.418827.00000 0004 0620 870XInstitute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Benes
- grid.418930.70000 0001 2299 1368Department of Cardiology, Institute for Clinical and Experimental Medicine - IKEM, Videnska 1958/9, 140 21 Prague 4, Czech Republic
| | - Ludek Cervenka
- grid.418930.70000 0001 2299 1368Department of Cardiology, Institute for Clinical and Experimental Medicine - IKEM, Videnska 1958/9, 140 21 Prague 4, Czech Republic ,grid.4491.80000 0004 1937 116XDepartment of Pathophysiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jiri Petrak
- grid.4491.80000 0004 1937 116XBIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Vojtech Melenovsky
- grid.418930.70000 0001 2299 1368Department of Cardiology, Institute for Clinical and Experimental Medicine - IKEM, Videnska 1958/9, 140 21 Prague 4, Czech Republic
| |
Collapse
|
17
|
Zhang N, Ye F, Zhou Y, Zhu W, Xie C, Zheng H, Chen H, Chen J, Xie X. Cardiac ankyrin repeat protein contributes to dilated cardiomyopathy and heart failure. FASEB J 2021; 35:e21488. [PMID: 33734499 DOI: 10.1096/fj.201902802rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 11/11/2022]
Abstract
Cardiac ankyrin repeat protein (CARP) is a cardiac-specific stress-response protein which exerts diverse effects to modulate cardiac remodeling in response to pathological stimuli. We examined the role of CARP in postnatal cardiac development and function under basal conditions in mice. Transgenic mice that selectively overexpressed CARP in heart (CARP Tg) exhibited dilated cardiac chambers, impaired heart function, and cardiac fibrosis as assessed by echocardiography and histological staining. Furthermore, the mice had a shorter lifespan and reduced survival rate in response to ischemic acute myocardial infarction. Immunofluorescence demonstrated the overexpressed CARP protein was predominantly accumulated in the nuclei of cardiomyocytes. Microarray analysis revealed that the nuclear localization of CARP was associated with the suppression of calcium-handling proteins. In vitro experiments revealed that CARP overexpression resulted in decreased cell contraction and calcium transient. In post-mortem cardiac specimens from patients with dilated cardiomyopathy and end-stage heart failure, CARP was significantly increased. Taken together, our data identified CARP as a crucial contributor in dilated cardiomyopathy and heart failure which was associated with its regulation of calcium-handling proteins.
Collapse
Affiliation(s)
- Na Zhang
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,School of Medicine, Hangzhou Normal University, Hangzhou, People's Republic of China
| | - Feiming Ye
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yu Zhou
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Wei Zhu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Cuiping Xie
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Haiqiong Zheng
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Han Chen
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jinghai Chen
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaojie Xie
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
18
|
Epigenetics and Heart Failure. Int J Mol Sci 2020; 21:ijms21239010. [PMID: 33260869 PMCID: PMC7729735 DOI: 10.3390/ijms21239010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
Epigenetics refers to changes in phenotypes without changes in genotypes. These changes take place in a number of ways, including via genomic DNA methylation, DNA interacting proteins, and microRNAs. The epigenome is the second dimension of the genome and it contains key information that is specific to every type of cell. Epigenetics is essential for many fundamental processes in biology, but its importance in the development and progression of heart failure, which is one of the major causes of morbidity and mortality worldwide, remains unclear. Our understanding of the underlying molecular mechanisms is incomplete. While epigenetics is one of the most innovative research areas in modern biology and medicine, compounds that directly target the epigenome, such as epidrugs, have not been well translated into therapies. This paper focuses on epigenetics in terms of genomic DNA methylation, such as 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) modifications. These appear to be more dynamic than previously anticipated and may underlie a wide variety of conditions, including heart failure. We also outline possible new strategies for the development of novel therapies.
Collapse
|
19
|
The p.(Cys150Tyr) variant in CSRP3 is associated with late-onset hypertrophic cardiomyopathy in heterozygous individuals. Eur J Med Genet 2020; 63:104079. [PMID: 33035702 DOI: 10.1016/j.ejmg.2020.104079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/06/2020] [Accepted: 09/29/2020] [Indexed: 01/25/2023]
Abstract
INTRODUCTION AND OBJECTIVES Up to 50% of patients with hypertrophic cardiomyopathy (HCM) show no disease-causing variants in genetic studies. Mutations in CSRP3 have been associated with HCM, but evidence supporting pathogenicity is inconclusive. In this study, we describe an HCM cohort with a missense variant in CSRP3 (p.Cys150Tyr) with supporting evidence for pathogenicity and a description of the associated phenotype. METHODS CSRP3 was sequenced in 6456 index cases with a diagnosis of HCM and in 5012 probands with other cardiomyopathies. In addition, 3372 index cases with hereditary cardiovascular disorders other than cardiomyopathies (mainly channelopathies and aortopathies) were used as controls. RESULTS The p.(Cys150Tyr) variant was identified in 11 unrelated individuals of the 6456 HCM probands, and it was not identified in patients with other cardiomyopathies (p < 0.0001) or in our control population (p < 0.0001). Ten of the index cases were heterozygous and one was homozygous. Homozygous had a more severe phenotype. Family screening identified 17 other carriers. Wild-type individuals showed no signs of disease. The mean age at diagnosis of affected individuals was 55 ± 13 years, and the mean left ventricular wall thickness was 18 ± 3 mm. The variant showed highly age-dependent penetrance. After a mean follow-up of 11 (±8) years, no adverse events were reported in any of the HCM patients. CONCLUSIONS The p.(Cys150Tyr) variant in CSRP3 causes late-onset and low risk form of hypertrophic cardiomyopathy in heterozygous carriers.
Collapse
|
20
|
Correcting an instance of synthetic lethality with a pro-survival sequence. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118734. [PMID: 32389645 DOI: 10.1016/j.bbamcr.2020.118734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 04/11/2020] [Accepted: 05/02/2020] [Indexed: 11/21/2022]
Abstract
A human cDNA encoding the LIM domain containing 194 amino acid cysteine and glycine rich protein 3 (CSRP3) was identified as a BAX suppressor in yeast and a pro-survival sequence that abrogated copper mediated regulated cell death (RCD). Yeast lacks a CSRP3 orthologue but it has four LIM sequences, namely RGA1, RGA2, LRG1 and PXL1. These are known regulators of stress responses yet their roles in RCD remain unknown. Given that LIMs interact with other LIMs, we ruled out the possibility that overexpressed yeast LIMs alone could prevent RCD and that CSRP3 functions by acting as a dominant regulator of yeast LIMs. Of interest was the discovery that even though yeast cells lacking the LIM encoding PXL1 had no overt growth defect, it was nevertheless supersensitive to the effects of sublethal levels of copper. Heterologous expression of human CSPR3 as well as the pro-survival 14-3-3 sequence corrected this copper supersensitivity. These results show that the pxl1∆-copper synthetic lethality is likely due to the induction of RCD. This differs from the prevailing model in which synthetic lethality occurs because of specific defects generated by the combined loss of two overlapping but non-essential functions.
Collapse
|
21
|
Putative Receptors for Gravity Sensing in Mammalian Cells: The Effects of Microgravity. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10062028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gravity is a constitutive force that influences life on Earth. It is sensed and translated into biochemical stimuli through the so called “mechanosensors”, proteins able to change their molecular conformation in order to amplify external cues causing several intracellular responses. Mechanosensors are widely represented in the human body with important structures such as otholiths in hair cells of vestibular system and statoliths in plants. Moreover, they are also present in the bone, where mechanical cues can cause bone resorption or formation and in muscle in which mechanical stimuli can increase the sensibility for mechanical stretch. In this review, we discuss the role of mechanosensors in two different conditions: normogravity and microgravity, emphasizing their emerging role in microgravity. Microgravity is a singular condition in which many molecular changes occur, strictly connected with the modified gravity force and free fall of bodies. Here, we first summarize the most important mechanosensors involved in normogravity and microgravity. Subsequently, we propose muscle LIM protein (MLP) and sirtuins as new actors in mechanosensing and signaling transduction under microgravity.
Collapse
|
22
|
Levin E, Leibinger M, Gobrecht P, Hilla A, Andreadaki A, Fischer D. Muscle LIM Protein Is Expressed in the Injured Adult CNS and Promotes Axon Regeneration. Cell Rep 2020; 26:1021-1032.e6. [PMID: 30673598 DOI: 10.1016/j.celrep.2018.12.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 09/05/2018] [Accepted: 12/05/2018] [Indexed: 01/01/2023] Open
Abstract
Muscle LIM protein (MLP) has long been regarded as a muscle-specific protein. Here, we report that MLP expression is induced in adult rat retinal ganglion cells (RGCs) upon axotomy, and its expression is correlated with their ability to regenerate injured axons. Specific knockdown of MLP in RGCs compromises axon regeneration, while overexpression in vivo facilitates optic nerve regeneration and regrowth of sensory neurons without affecting neuronal survival. MLP accumulates in the cell body, the nucleus, and in axonal growth cones, which are significantly enlarged by its overexpression. Only the MLP fraction in growth cones is relevant for promoting axon extension. Additional data suggest that MLP acts as an actin cross-linker, thereby facilitating filopodia formation and increasing growth cone motility. Thus, MLP-mediated effects on actin could become a therapeutic strategy for promoting nerve repair.
Collapse
Affiliation(s)
- Evgeny Levin
- Division of Experimental Neurology, Medical Faculty, Heinrich Heine University, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| | - Marco Leibinger
- Department of Cell Physiology, Ruhr University of Bochum, Universitätsstraße 150, 44780 Bochum, Germany; Division of Experimental Neurology, Medical Faculty, Heinrich Heine University, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| | - Philipp Gobrecht
- Department of Cell Physiology, Ruhr University of Bochum, Universitätsstraße 150, 44780 Bochum, Germany; Division of Experimental Neurology, Medical Faculty, Heinrich Heine University, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| | - Alexander Hilla
- Department of Cell Physiology, Ruhr University of Bochum, Universitätsstraße 150, 44780 Bochum, Germany; Division of Experimental Neurology, Medical Faculty, Heinrich Heine University, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| | - Anastasia Andreadaki
- Department of Cell Physiology, Ruhr University of Bochum, Universitätsstraße 150, 44780 Bochum, Germany; Division of Experimental Neurology, Medical Faculty, Heinrich Heine University, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| | - Dietmar Fischer
- Department of Cell Physiology, Ruhr University of Bochum, Universitätsstraße 150, 44780 Bochum, Germany; Division of Experimental Neurology, Medical Faculty, Heinrich Heine University, Merowingerplatz 1a, 40225 Düsseldorf, Germany.
| |
Collapse
|
23
|
Cui C, Han S, Tang S, He H, Shen X, Zhao J, Chen Y, Wei Y, Wang Y, Zhu Q, Li D, Yin H. The Autophagy Regulatory Molecule CSRP3 Interacts with LC3 and Protects Against Muscular Dystrophy. Int J Mol Sci 2020; 21:ijms21030749. [PMID: 31979369 PMCID: PMC7037376 DOI: 10.3390/ijms21030749] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 12/22/2022] Open
Abstract
CSRP3/MLP (cysteine-rich protein 3/muscle Lim protein), a member of the cysteine-rich protein family, is a muscle-specific LIM-only factor specifically expressed in skeletal muscle. CSRP3 is critical in maintaining the structure and function of normal muscle. To investigate the mechanism of disease in CSRP3 myopathy, we performed siRNA-mediated CSRP3 knockdown in chicken primary myoblasts. CSRP3 silencing resulted in the down-regulation of the expression of myogenic genes and the up-regulation of atrophy-related gene expressions. We found that CSRP3 interacted with LC3 protein to promote the formation of autophagosomes during autophagy. CSRP3-silencing impaired myoblast autophagy, as evidenced by inhibited autophagy-related ATG5 and ATG7 mRNA expression levels, and inhibited LC3II and Beclin-1 protein accumulation. In addition, impaired autophagy in CSRP3-silenced cells resulted in increased sensitivity to apoptosis cell death. CSRP3-silenced cells also showed increased caspase-3 and caspase-9 cleavage. Moreover, apoptosis induced by CSRP3 silencing was alleviated after autophagy activation. Together, these results indicate that CSRP3 promotes the correct formation of autophagosomes through its interaction with LC3 protein, which has an important role in skeletal muscle remodeling and maintenance.
Collapse
|
24
|
Tang X, Pan L, Zhao S, Dai F, Chao M, Jiang H, Li X, Lin Z, Huang Z, Meng G, Wang C, Chen C, Liu J, Wang X, Ferro A, Wang H, Chen H, Gao Y, Lu Q, Xie L, Han Y, Ji Y. SNO-MLP (S-Nitrosylation of Muscle LIM Protein) Facilitates Myocardial Hypertrophy Through TLR3 (Toll-Like Receptor 3)-Mediated RIP3 (Receptor-Interacting Protein Kinase 3) and NLRP3 (NOD-Like Receptor Pyrin Domain Containing 3) Inflammasome Activation. Circulation 2020; 141:984-1000. [PMID: 31902237 DOI: 10.1161/circulationaha.119.042336] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND S-nitrosylation (SNO), a prototypic redox-based posttranslational modification, is involved in the pathogenesis of cardiovascular disease. The aim of this study was to determine the role of SNO of MLP (muscle LIM protein) in myocardial hypertrophy, as well as the mechanism by which SNO-MLP modulates hypertrophic growth in response to pressure overload. METHODS Myocardial samples from patients and animal models exhibiting myocardial hypertrophy were examined for SNO-MLP level using biotin-switch methods. SNO sites were further identified through liquid chromatography-tandem mass spectrometry. Denitrosylation of MLP by the mutation of nitrosylation sites or overexpression of S-nitrosoglutathione reductase was used to analyze the contribution of SNO-MLP in myocardial hypertrophy. Downstream effectors of SNO-MLP were screened through mass spectrometry and confirmed by coimmunoprecipitation. Recruitment of TLR3 (Toll-like receptor 3) by SNO-MLP in myocardial hypertrophy was examined in TLR3 small interfering RNA-transfected neonatal rat cardiomyocytes and in a TLR3 knockout mouse model. RESULTS SNO-MLP level was significantly higher in hypertrophic myocardium from patients and in spontaneously hypertensive rats and mice subjected to transverse aortic constriction. The level of SNO-MLP also increased in angiotensin II- or phenylephrine-treated neonatal rat cardiomyocytes. S-nitrosylated site of MLP at cysteine 79 was identified by liquid chromatography-tandem mass spectrometry and confirmed in neonatal rat cardiomyocytes. Mutation of cysteine 79 significantly reduced hypertrophic growth in angiotensin II- or phenylephrine-treated neonatal rat cardiomyocytes and transverse aortic constriction mice. Reducing SNO-MLP level by overexpression of S-nitrosoglutathione reductase greatly attenuated myocardial hypertrophy. Mechanistically, SNO-MLP stimulated TLR3 binding to MLP in response to hypertrophic stimuli, and disrupted this interaction by downregulating TLR3-attenuated myocardial hypertrophy. SNO-MLP also increased the complex formation between TLR3 and RIP3 (receptor-interacting protein kinase 3). This interaction in turn induced NLRP3 (nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3) inflammasome activation, thereby promoting the development of myocardial hypertrophy. CONCLUSIONS Our findings revealed a key role of SNO-MLP in myocardial hypertrophy and demonstrated TLR3-mediated RIP3 and NLRP3 inflammasome activation as the downstream signaling pathway, which may represent a therapeutic target for myocardial hypertrophy and heart failure.
Collapse
Affiliation(s)
- Xin Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Lihong Pan
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Shuang Zhao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Feiyue Dai
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Menglin Chao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Hong Jiang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Xuesong Li
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Zhe Lin
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Zhengrong Huang
- Department of Cardiology, the First Affiliated Hospital of Xiamen University, China (Z.H.)
| | - Guoliang Meng
- Nanjing Medical University, Nanjing, China (G.M.).,Department of Pharmacology, School of Pharmacy, Nantong University, China (G.M.)
| | - Chun Wang
- Department of Geriatrics, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, China (C.W.)
| | - Chan Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China (C.C., J.L.)
| | - Jin Liu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China (C.C., J.L.)
| | - Xin Wang
- Faculty of Biology, Medicine and Health, the University of Manchester, United Kingdom (X.W.)
| | - Albert Ferro
- Cardiovascular Clinical Pharmacology, British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College London, United Kingdom (A.F.)
| | - Hong Wang
- Department of Pharmacology, Lewis Kats School of Medicine, Temple University, Philadelphia, PA (H.W.)
| | - Hongshan Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Yuanqing Gao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Qiulun Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Liping Xie
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Yi Han
- Department of Geriatrics, First Affiliated Hospital of Nanjing Medical University, China (Y.H.)
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.).,State Key Laboratory of Reproductive Medicine (Y.J.)
| |
Collapse
|
25
|
Cai W, Zhang J, de Lange WJ, Gregorich ZR, Karp H, Farrell ET, Mitchell SD, Tucholski T, Lin Z, Biermann M, McIlwain SJ, Ralphe JC, Kamp TJ, Ge Y. An Unbiased Proteomics Method to Assess the Maturation of Human Pluripotent Stem Cell-Derived Cardiomyocytes. Circ Res 2019; 125:936-953. [PMID: 31573406 PMCID: PMC6852699 DOI: 10.1161/circresaha.119.315305] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022]
Abstract
RATIONALE Human pluripotent stem cell (hPSC)-derived cardiomyocytes exhibit the properties of fetal cardiomyocytes, which limits their applications. Various methods have been used to promote maturation of hPSC-cardiomyocytes; however, there is a lack of an unbiased and comprehensive method for accurate assessment of the maturity of hPSC-cardiomyocytes. OBJECTIVE We aim to develop an unbiased proteomics strategy integrating high-throughput top-down targeted proteomics and bottom-up global proteomics for the accurate and comprehensive assessment of hPSC-cardiomyocyte maturation. METHODS AND RESULTS Utilizing hPSC-cardiomyocytes from early- and late-stage 2-dimensional monolayer culture and 3-dimensional engineered cardiac tissue, we demonstrated the high reproducibility and reliability of a top-down proteomics method, which enabled simultaneous quantification of contractile protein isoform expression and associated post-translational modifications. This method allowed for the detection of known maturation-associated contractile protein alterations and, for the first time, identified contractile protein post-translational modifications as promising new markers of hPSC-cardiomyocytes maturation. Most notably, decreased phosphorylation of α-tropomyosin was found to be associated with hPSC-cardiomyocyte maturation. By employing a bottom-up global proteomics strategy, we identified candidate maturation-associated markers important for sarcomere organization, cardiac excitability, and Ca2+ homeostasis. In particular, upregulation of myomesin 1 and transmembrane 65 was associated with hPSC-cardiomyocyte maturation and validated in cardiac development, making these promising markers for assessing maturity of hPSC-cardiomyocytes. We have further validated α-actinin isoforms, phospholamban, dystrophin, αB-crystallin, and calsequestrin 2 as novel maturation-associated markers, in the developing mouse cardiac ventricles. CONCLUSIONS We established an unbiased proteomics method that can provide accurate and specific assessment of the maturity of hPSC-cardiomyocytes and identified new markers of maturation. Furthermore, this integrated proteomics strategy laid a strong foundation for uncovering the molecular pathways involved in cardiac development and disease using hPSC-cardiomyocytes.
Collapse
Affiliation(s)
- Wenxuan Cai
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jianhua Zhang
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Willem J. de Lange
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachery R. Gregorich
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Hannah Karp
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Emily T. Farrell
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Stanford D. Mitchell
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Trisha Tucholski
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ziqing Lin
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
- Human Proteomics Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mitch Biermann
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sean J. McIlwain
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - J. Carter Ralphe
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Timothy J. Kamp
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ying Ge
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
- Human Proteomics Program, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
26
|
Oudot C, Gomes A, Nicolas V, Le Gall M, Chaffey P, Broussard C, Calamita G, Mastrodonato M, Gena P, Perfettini JL, Hamelin J, Lemoine A, Fischmeister R, Vieira HL, Santos CN, Brenner C. CSRP3 mediates polyphenols-induced cardioprotection in hypertension. J Nutr Biochem 2019; 66:29-42. [DOI: 10.1016/j.jnutbio.2019.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 11/23/2018] [Accepted: 01/02/2019] [Indexed: 12/16/2022]
|
27
|
Mass Spectrometry Based Comparative Proteomics Using One Dimensional and Two Dimensional SDS-PAGE of Rat Atria Induced with Obstructive Sleep Apnea. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:541-561. [DOI: 10.1007/978-3-030-15950-4_32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
28
|
Bond AR, Iacobazzi D, Abdul-Ghani S, Ghorbel M, Heesom K, Wilson M, Gillett C, George SJ, Caputo M, Suleiman S, Tulloh RMR. Changes in contractile protein expression are linked to ventricular stiffness in infants with pulmonary hypertension or right ventricular hypertrophy due to congenital heart disease. Open Heart 2018; 5:e000716. [PMID: 29344379 PMCID: PMC5761287 DOI: 10.1136/openhrt-2017-000716] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/15/2017] [Accepted: 11/18/2017] [Indexed: 12/12/2022] Open
Abstract
Background The right ventricle (RV) is not designed to sustain high pressure leading to failure. There are no current medications to help RV contraction, so further information is required on adaption of the RV to such hypertension. Methods The Right Ventricle in Children (RVENCH) study assessed infants with congenital heart disease undergoing cardiac surgery with hypertensive RV. Clinical and echocardiographic data were recorded, and samples of RV were taken from matched infants, analysed for proteomics and compared between pathologies and with clinical and echocardiographic outcome data. Results Those with tetralogy of Fallot (TOF) were significantly more cyanosed than those with ventricular septal defect (median oxygen saturation 83% vs 98%, P=0.0038), had significantly stiffer RV (tricuspid E wave/A wave ratio 1.95 vs 0.84, P=0.009) and had most had restrictive physiology. Gene ontology in TOF, with enrichment analysis, demonstrated significant increase in proteins of contractile mechanisms and those of calmodulin, actin binding and others associated with contractility than inventricular septal defect. Structural proteins were also found to be higher in association with sarcomeric function: Z-disc, M-Band and thin-filament proteins. Remaining proteins associated with actin binding, calcium signalling and myocyte cytoskeletal development. Phosphopeptide enrichment led to higher levels of calcium signalling proteins in TOF. Conclusion This is the first demonstration that those with an RV, which is stiff and hypertensive in TOF, have a range of altered proteins, often in calcium signalling pathways. Information about these alterations might guide treatment options both in terms of individualised therapy or inotropic support for the Right ventricle when hypertensive due to pulmoanry hypertension or congenital heart disease.
Collapse
Affiliation(s)
- Andrew R Bond
- Clinical Sciences, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | - Dominga Iacobazzi
- Clinical Sciences, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | - Safa Abdul-Ghani
- Clinical Sciences, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | - Mohammed Ghorbel
- Clinical Sciences, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | - Kate Heesom
- Proteomics Facility, University of Bristol, Bristol, UK
| | | | | | - Sarah J George
- Clinical Sciences, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | - Massimo Caputo
- Clinical Sciences, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK.,Department of Congenital Heart Disease, King David Building, Bristol, UK
| | - Saadeh Suleiman
- Clinical Sciences, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | - Robert M R Tulloh
- Clinical Sciences, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK.,Department of Congenital Heart Disease, King David Building, Bristol, UK
| |
Collapse
|
29
|
Muscle Lim Protein and myosin binding protein C form a complex regulating muscle differentiation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2308-2321. [DOI: 10.1016/j.bbamcr.2017.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 08/09/2017] [Accepted: 08/30/2017] [Indexed: 01/10/2023]
|
30
|
Ehsan M, Jiang H, L Thomson K, Gehmlich K. When signalling goes wrong: pathogenic variants in structural and signalling proteins causing cardiomyopathies. J Muscle Res Cell Motil 2017; 38:303-316. [PMID: 29119312 PMCID: PMC5742121 DOI: 10.1007/s10974-017-9487-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/28/2017] [Indexed: 12/20/2022]
Abstract
Cardiomyopathies are a diverse group of cardiac disorders with distinct phenotypes, depending on the proteins and pathways affected. A substantial proportion of cardiomyopathies are inherited and those will be the focus of this review article. With the wide application of high-throughput sequencing in the practice of clinical genetics, the roles of novel genes in cardiomyopathies are recognised. Here, we focus on a subgroup of cardiomyopathy genes [TTN, FHL1, CSRP3, FLNC and PLN, coding for Titin, Four and a Half LIM domain 1, Muscle LIM Protein, Filamin C and Phospholamban, respectively], which, despite their diverse biological functions, all have important signalling functions in the heart, suggesting that disturbances in signalling networks can contribute to cardiomyopathies.
Collapse
Affiliation(s)
- Mehroz Ehsan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - He Jiang
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Kate L Thomson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK.
| |
Collapse
|
31
|
Channaveerappa D, Lux JC, Wormwood KL, Heintz TA, McLerie M, Treat JA, King H, Alnasser D, Goodrow RJ, Ballard G, Decker R, Darie CC, Panama BK. Atrial electrophysiological and molecular remodelling induced by obstructive sleep apnoea. J Cell Mol Med 2017; 21:2223-2235. [PMID: 28402037 PMCID: PMC5571519 DOI: 10.1111/jcmm.13145] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 01/31/2017] [Indexed: 12/19/2022] Open
Abstract
Obstructive sleep apnoea (OSA) affects 9-24% of the adult population. OSA is associated with atrial disease, including atrial enlargement, fibrosis and arrhythmias. Despite the link between OSA and cardiac disease, the molecular changes in the heart which occur with OSA remain elusive. To study OSA-induced cardiac changes, we utilized a recently developed rat model which closely recapitulates the characteristics of OSA. Male Sprague Dawley rats, aged 50-70 days, received surgically implanted tracheal balloons which were inflated to cause transient airway obstructions. Rats were given 60 apnoeas per hour of either 13 sec. (moderate apnoea) or 23 sec. (severe apnoea), 8 hrs per day for 2 weeks. Controls received implants, but no inflations were made. Pulse oximetry measurements were taken at regular intervals, and post-apnoea ECGs were recorded. Rats had longer P wave durations and increased T wave amplitudes following chronic OSA. Proteomic analysis of the atrial tissue homogenates revealed that three of the nine enzymes in glycolysis, and two proteins related to oxidative phosphorylation, were down regulated in the severe apnoea group. Several sarcomeric and pro-hypertrophic proteins were also up regulated with OSA. Chronic OSA causes proteins changes in the atria which suggest impairment of energy metabolism and enhancement of hypertrophy.
Collapse
Affiliation(s)
- Devika Channaveerappa
- Biochemistry and Proteomics GroupDepartment of Chemistry and Biomolecular ScienceClarkson UniversityPotsdamNYUSA
| | - Jacob C. Lux
- Department of Experimental CardiologyMasonic Medical Research LaboratoryUticaNYUSA
| | - Kelly L. Wormwood
- Biochemistry and Proteomics GroupDepartment of Chemistry and Biomolecular ScienceClarkson UniversityPotsdamNYUSA
| | - Timothy A. Heintz
- Department of Experimental CardiologyMasonic Medical Research LaboratoryUticaNYUSA
| | - Meredith McLerie
- Department of Experimental CardiologyMasonic Medical Research LaboratoryUticaNYUSA
| | - Jacqueline A. Treat
- Department of Experimental CardiologyMasonic Medical Research LaboratoryUticaNYUSA
| | - Hannah King
- Department of Experimental CardiologyMasonic Medical Research LaboratoryUticaNYUSA
| | - Donia Alnasser
- Department of Experimental CardiologyMasonic Medical Research LaboratoryUticaNYUSA
| | - Robert J. Goodrow
- Department of Experimental CardiologyMasonic Medical Research LaboratoryUticaNYUSA
| | - Glenn Ballard
- Electrical Engineering TechnologyMohawk Valley Community CollegeUticaNYUSA
| | - Robert Decker
- Electrical Engineering TechnologyMohawk Valley Community CollegeUticaNYUSA
| | - Costel C. Darie
- Biochemistry and Proteomics GroupDepartment of Chemistry and Biomolecular ScienceClarkson UniversityPotsdamNYUSA
| | - Brian K. Panama
- Department of Experimental CardiologyMasonic Medical Research LaboratoryUticaNYUSA
| |
Collapse
|
32
|
Nociceptive DRG neurons express muscle lim protein upon axonal injury. Sci Rep 2017; 7:643. [PMID: 28377582 PMCID: PMC5428558 DOI: 10.1038/s41598-017-00590-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/03/2017] [Indexed: 01/21/2023] Open
Abstract
Muscle lim protein (MLP) has long been regarded as a cytosolic and nuclear muscular protein. Here, we show that MLP is also expressed in a subpopulation of adult rat dorsal root ganglia (DRG) neurons in response to axonal injury, while the protein was not detectable in naïve cells. Detailed immunohistochemical analysis of L4/L5 DRG revealed ~3% of MLP-positive neurons 2 days after complete sciatic nerve crush and maximum ~10% after 4–14 days. Similarly, in mixed cultures from cervical, thoracic, lumbar and sacral DRG ~6% of neurons were MLP-positive after 2 days and maximal 17% after 3 days. In both, histological sections and cell cultures, the protein was detected in the cytosol and axons of small diameter cells, while the nucleus remained devoid. Moreover, the vast majority could not be assigned to any of the well characterized canonical DRG subpopulations at 7 days after nerve injury. However, further analysis in cell culture revealed that the largest population of MLP expressing cells originated from non-peptidergic IB4-positive nociceptive neurons, which lose their ability to bind the lectin upon axotomy. Thus, MLP is mostly expressed in a subset of axotomized nociceptive neurons and can be used as a novel marker for this population of cells.
Collapse
|
33
|
Straubinger J, Boldt K, Kuret A, Deng L, Krattenmacher D, Bork N, Desch M, Feil R, Feil S, Nemer M, Ueffing M, Ruth P, Just S, Lukowski R. Amplified pathogenic actions of angiotensin II in cysteine-rich LIM-only protein 4-negative mouse hearts. FASEB J 2017; 31:1620-1638. [PMID: 28138039 DOI: 10.1096/fj.201601186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022]
Abstract
LIM domain proteins have been identified as essential modulators of cardiac biology and pathology; however, it is unclear which role the cysteine-rich LIM-only protein (CRP)4 plays in these processes. In studying CRP4 mutant mice, we found that their hearts developed normally, but lack of CRP4 exaggerated multiple parameters of the cardiac stress response to the neurohormone angiotensin II (Ang II). Aiming to dissect the molecular details, we found a link between CRP4 and the cardioprotective cGMP pathway, as well as a multiprotein complex comprising well-known hypertrophy-associated factors. Significant enrichment of the cysteine-rich intestinal protein (CRIP)1 in murine hearts lacking CRP4, as well as severe cardiac defects and premature death of CRIP1 and CRP4 morphant zebrafish embryos, further support the notion that depleting CRP4 is incompatible with a proper cardiac development and function. Together, amplified Ang II signaling identified CRP4 as a novel antiremodeling factor regulated, at least to some extent, by cardiac cGMP.-Straubinger, J., Boldt, K., Kuret, A., Deng, L., Krattenmacher, D., Bork, N., Desch, M., Feil, R., Feil, S., Nemer, M., Ueffing, M., Ruth, P., Just, S., Lukowski, R. Amplified pathogenic actions of angiotensin II in cysteine-rich LIM-only protein 4 negative mouse hearts.
Collapse
Affiliation(s)
- Julia Straubinger
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Karsten Boldt
- Institute for Ophthalmic Research, Molecular Biology of Retinal Degenerations and Medical Proteome Center, University of Tübingen, Tübingen, Germany
| | - Anna Kuret
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Lisa Deng
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Diana Krattenmacher
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Nadja Bork
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Matthias Desch
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Robert Feil
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany; and
| | - Susanne Feil
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany; and
| | - Mona Nemer
- Laboratory of Cardiac Development and Differentiation, Department of Biochemistry, Immunology, and Microbiology, University of Ottawa, Ottawa, Ontario, Canada
| | - Marius Ueffing
- Institute for Ophthalmic Research, Molecular Biology of Retinal Degenerations and Medical Proteome Center, University of Tübingen, Tübingen, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Steffen Just
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany;
| |
Collapse
|
34
|
Rashid MM, Runci A, Russo MA, Tafani M. Muscle Lim Protein (MLP)/CSRP3 at the crossroad between mechanotransduction and autophagy. Cell Death Dis 2015; 6:e1940. [PMID: 26492373 PMCID: PMC4632324 DOI: 10.1038/cddis.2015.308] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- M M Rashid
- Department of Experimental Medicine, University of Rome, Sapienza, Rome, Italy
| | - A Runci
- Department of Experimental Medicine, University of Rome, Sapienza, Rome, Italy
| | - M A Russo
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele, Rome, Italy
| | - M Tafani
- Department of Experimental Medicine, University of Rome, Sapienza, Rome, Italy
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele, Rome, Italy
| |
Collapse
|
35
|
Rashid MM, Runci A, Polletta L, Carnevale I, Morgante E, Foglio E, Arcangeli T, Sansone L, Russo MA, Tafani M. Muscle LIM protein/CSRP3: a mechanosensor with a role in autophagy. Cell Death Discov 2015; 1:15014. [PMID: 27551448 PMCID: PMC4981024 DOI: 10.1038/cddiscovery.2015.14] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 06/11/2015] [Accepted: 06/12/2015] [Indexed: 12/12/2022] Open
Abstract
Muscle LIM protein (MLP) is a microtubule-associated protein expressed in cardiac and muscle tissues that belongs to the cysteine-rich protein (CSRP/CRP) family. MLP has a central role during muscle development and for architectural maintenance of muscle cells. However, muscle cells rely on autophagy during differentiation and for structural maintenance. To study the role of MLP in autophagy, we have used C2C12 mouse myoblasts silenced or overexpressing MLP. Our results show that MLP contributes to the correct autophagosome formation and flux by interacting with LC3 as demonstrated by co-immunoprecipitation and PLA assay. In fact, MLP silencing results in decreased LC3-II staining and absent degradation of long-lived proteins. Moreover, MLP silencing impaired myoblasts differentiation as measured by decreased expression of MyoD1, MyoG1 and myosin heavy chain. Ultrastructural analysis revealed the presence of large empty autophagosomes in myoblasts and multimembranous structures in myotubes from MLP-silenced clones. Impaired autophagy in MLP-silenced cells resulted in increased susceptibility to apoptotic cell death. In fact, treatment of MLP-silenced C2C12 myoblasts and myotubes with staurosporine resulted in increased caspase-3 and PARP cleavage as well as increased percentage of cell death. In conclusion, we propose that MLP regulates autophagy during muscle cell differentiation or maintenance through a mechanism involving MLP/LC3-II interaction and correct autophagosome formation.
Collapse
Affiliation(s)
- M M Rashid
- Department of Experimental Medicine, University of Rome , Sapienza, Rome, Italy
| | - A Runci
- Department of Experimental Medicine, University of Rome , Sapienza, Rome, Italy
| | - L Polletta
- Department of Experimental Medicine, University of Rome , Sapienza, Rome, Italy
| | - I Carnevale
- Department of Experimental Medicine, University of Rome , Sapienza, Rome, Italy
| | - E Morgante
- Department of Experimental Medicine, University of Rome , Sapienza, Rome, Italy
| | - E Foglio
- Department of Experimental Medicine, University of Rome , Sapienza, Rome, Italy
| | - T Arcangeli
- Department of Experimental Medicine, University of Rome , Sapienza, Rome, Italy
| | - L Sansone
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele , Rome, Italy
| | - M A Russo
- Consorzio MEBIC, San Raffaele University , Rome, Italy
| | - M Tafani
- Department of Experimental Medicine, University of Rome, Sapienza, Rome, Italy; Department of Cellular and Molecular Pathology, IRCCS San Raffaele, Rome, Italy
| |
Collapse
|
36
|
Vafiadaki E, Arvanitis DA, Sanoudou D. Muscle LIM Protein: Master regulator of cardiac and skeletal muscle functions. Gene 2015; 566:1-7. [PMID: 25936993 PMCID: PMC6660132 DOI: 10.1016/j.gene.2015.04.077] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 04/21/2015] [Accepted: 04/27/2015] [Indexed: 12/17/2022]
Abstract
Muscle LIM Protein (MLP) has emerged as a key regulator of striated muscle physiology and pathophysiology. Mutations in cysteine and glycine-rich protein 3 (CSRP3), the gene encoding MLP, are causative of human cardiomyopathies, whereas altered expression patterns are observed in human failing heart and skeletal myopathies. In vitro and in vivo evidences reveal a complex and diverse functional role of MLP in striated muscle, which is determined by its multiple interacting partners and subcellular distribution. Experimental evidence suggests that MLP is implicated in both myogenic differentiation and myocyte cytoarchitecture, although the full spectrum of its intracellular roles still unfolds.
Collapse
Affiliation(s)
- Elizabeth Vafiadaki
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Greece
| | - Demetrios A Arvanitis
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Greece
| | - Despina Sanoudou
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Greece; 4th Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
37
|
Buyandelger B, Mansfield C, Kostin S, Choi O, Roberts AM, Ware JS, Mazzarotto F, Pesce F, Buchan R, Isaacson RL, Vouffo J, Gunkel S, Knöll G, McSweeney SJ, Wei H, Perrot A, Pfeiffer C, Toliat MR, Ilieva K, Krysztofinska E, López-Olañeta MM, Gómez-Salinero JM, Schmidt A, Ng KE, Teucher N, Chen J, Teichmann M, Eilers M, Haverkamp W, Regitz-Zagrosek V, Hasenfuss G, Braun T, Pennell DJ, Gould I, Barton PJR, Lara-Pezzi E, Schäfer S, Hübner N, Felkin LE, O'Regan DP, Brand T, Milting H, Nürnberg P, Schneider MD, Prasad S, Petretto E, Knöll R. ZBTB17 (MIZ1) Is Important for the Cardiac Stress Response and a Novel Candidate Gene for Cardiomyopathy and Heart Failure. ACTA ACUST UNITED AC 2015; 8:643-52. [PMID: 26175529 DOI: 10.1161/circgenetics.113.000690] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 07/02/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Mutations in sarcomeric and cytoskeletal proteins are a major cause of hereditary cardiomyopathies, but our knowledge remains incomplete as to how the genetic defects execute their effects. METHODS AND RESULTS We used cysteine and glycine-rich protein 3, a known cardiomyopathy gene, in a yeast 2-hybrid screen and identified zinc-finger and BTB domain-containing protein 17 (ZBTB17) as a novel interacting partner. ZBTB17 is a transcription factor that contains the peak association signal (rs10927875) at the replicated 1p36 cardiomyopathy locus. ZBTB17 expression protected cardiac myocytes from apoptosis in vitro and in a mouse model with cardiac myocyte-specific deletion of Zbtb17, which develops cardiomyopathy and fibrosis after biomechanical stress. ZBTB17 also regulated cardiac myocyte hypertrophy in vitro and in vivo in a calcineurin-dependent manner. CONCLUSIONS We revealed new functions for ZBTB17 in the heart, a transcription factor that may play a role as a novel cardiomyopathy gene.
Collapse
|
38
|
Levin E, Leibinger M, Andreadaki A, Fischer D. Neuronal expression of muscle LIM protein in postnatal retinae of rodents. PLoS One 2014; 9:e100756. [PMID: 24945278 PMCID: PMC4063954 DOI: 10.1371/journal.pone.0100756] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/30/2014] [Indexed: 01/22/2023] Open
Abstract
Muscle LIM protein (MLP) is a member of the cysteine rich protein family and has so far been regarded as a muscle-specific protein that is mainly involved in myogenesis and the organization of cytoskeletal structure in myocytes, respectively. The current study demonstrates for the first time that MLP expression is not restricted to muscle tissue, but is also found in the rat naive central nervous system. Using quantitative PCR, Western blot and immunohistochemical analyses we detected MLP in the postnatal rat retina, specifically in the somas and dendritic arbors of cholinergic amacrine cells (AC) of the inner nuclear layer and the ganglion cell layer (displaced AC). Induction of MLP expression started at embryonic day 20 and peaked between postnatal days 7 and 14. It subsequently decreased again to non-detectable protein levels after postnatal day 28. MLP was identified in the cytoplasm and dendrites but not in the nucleus of AC. Thus, retinal MLP expression correlates with the morphologic and functional development of cholinergic AC, suggesting a potential role of this protein in postnatal maturation and making MLP a suitable marker for these neurons.
Collapse
Affiliation(s)
- Evgeny Levin
- Division of Experimental Neurology, Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Marco Leibinger
- Division of Experimental Neurology, Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Anastasia Andreadaki
- Division of Experimental Neurology, Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Dietmar Fischer
- Division of Experimental Neurology, Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
39
|
Human muscle LIM protein dimerizes along the actin cytoskeleton and cross-links actin filaments. Mol Cell Biol 2014; 34:3053-65. [PMID: 24934443 DOI: 10.1128/mcb.00651-14] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The muscle LIM protein (MLP) is a nucleocytoplasmic shuttling protein playing important roles in the regulation of myocyte remodeling and adaptation to hypertrophic stimuli. Missense mutations in human MLP or its ablation in transgenic mice promotes cardiomyopathy and heart failure. The exact function(s) of MLP in the cytoplasmic compartment and the underlying molecular mechanisms remain largely unknown. Here, we provide evidence that MLP autonomously binds to, stabilizes, and bundles actin filaments (AFs) independently of calcium and pH. Using total internal reflection fluorescence microscopy, we have shown how MLP cross-links actin filaments into both unipolar and mixed-polarity bundles. Quantitative analysis of the actin cytoskeleton configuration confirmed that MLP substantially promotes actin bundling in live myoblasts. In addition, bimolecular fluorescence complementation (BiFC) assays revealed MLP self-association. Remarkably, BiFC complexes mostly localize along actin filament-rich structures, such as stress fibers and sarcomeres, supporting a functional link between MLP self-association and actin cross-linking. Finally, we have demonstrated that MLP self-associates through its N-terminal LIM domain, whereas it binds to AFs through its C-terminal LIM domain. Together our data support that MLP contributes to the maintenance of cardiomyocyte cytoarchitecture by a mechanism involving its self-association and actin filament cross-linking.
Collapse
|
40
|
Vafiadaki E, Arvanitis DA, Papalouka V, Terzis G, Roumeliotis TI, Spengos K, Garbis SD, Manta P, Kranias EG, Sanoudou D. Muscle lim protein isoform negatively regulates striated muscle actin dynamics and differentiation. FEBS J 2014; 281:3261-79. [PMID: 24860983 DOI: 10.1111/febs.12859] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 05/14/2014] [Accepted: 05/22/2014] [Indexed: 11/28/2022]
Abstract
Muscle lim protein (MLP) has emerged as a critical regulator of striated muscle physiology and pathophysiology. Mutations in cysteine and glycine-rich protein 3 (CSRP3), the gene encoding MLP, have been directly associated with human cardiomyopathies, whereas aberrant expression patterns are reported in human cardiac and skeletal muscle diseases. Increasing evidence suggests that MLP has an important role in both myogenic differentiation and myocyte cytoarchitecture, although the full spectrum of its intracellular roles has not been delineated. We report the discovery of an alternative splice variant of MLP, designated as MLP-b, showing distinct expression in neuromuscular disease and direct roles in actin dynamics and muscle differentiation. This novel isoform originates by alternative splicing of exons 3 and 4. At the protein level, it contains the N-terminus first half LIM domain of MLP and a unique sequence of 22 amino acids. Physiologically, it is expressed during early differentiation, whereas its overexpression reduces C2C12 differentiation and myotube formation. This may be mediated through its inhibition of MLP/cofilin-2-mediated F-actin dynamics. In differentiated striated muscles, MLP-b localizes to the sarcomeres and binds directly to Z-disc components, including α-actinin, T-cap and MLP. The findings of the present study unveil a novel player in muscle physiology and pathophysiology that is implicated in myogenesis as a negative regulator of myotube formation, as well as in differentiated striated muscles as a contributor to sarcomeric integrity.
Collapse
Affiliation(s)
- Elizabeth Vafiadaki
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kaushik G, Engler AJ. From stem cells to cardiomyocytes: the role of forces in cardiac maturation, aging, and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 126:219-42. [PMID: 25081620 DOI: 10.1016/b978-0-12-394624-9.00009-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Stem cell differentiation into a variety of lineages is known to involve signaling from the extracellular niche, including from the physical properties of that environment. What regulates stem cell responses to these cues is there ability to activate different mechanotransductive pathways. Here, we will review the structures and pathways that regulate stem cell commitment to a cardiomyocyte lineage, specifically examining proteins within muscle sarcomeres, costameres, and intercalated discs. Proteins within these structures stretch, inducing a change in their phosphorylated state or in their localization to initiate different signals. We will also put these changes in the context of stem cell differentiation into cardiomyocytes, their subsequent formation of the chambered heart, and explore negative signaling that occurs during disease.
Collapse
Affiliation(s)
- Gaurav Kaushik
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
42
|
Clark KA, Kadrmas JL. Drosophila melanogaster muscle LIM protein and alpha-actinin function together to stabilize muscle cytoarchitecture: a potential role for Mlp84B in actin-crosslinking. Cytoskeleton (Hoboken) 2013; 70:304-16. [PMID: 23606669 PMCID: PMC3716849 DOI: 10.1002/cm.21106] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 03/04/2013] [Accepted: 03/06/2013] [Indexed: 02/06/2023]
Abstract
Stabilization of tissue architecture during development and growth is essential to maintain structural integrity. Because of its contractile nature, muscle is especially susceptible to physiological stresses, and has multiple mechanisms to maintain structural integrity. The Drosophila melanogaster Muscle LIM Protein (MLP), Mlp84B, participates in muscle maintenance, yet its precise mechanism of action is still controversial. Through a candidate approach, we identified α-actinin as a protein that functions with Mlp84B to ensure muscle integrity. α-actinin RNAi animals die primarily as pupae, and Mlp84B RNAi animals are adult viable. RNAi knockdown of Mlp84B and α-actinin together produces synergistic early larval lethality and destabilization of Z-line structures. We recapitulated these phenotypes using combinations of traditional loss-of-function alleles and single-gene RNAi. We observe that Mlp84B induces the formation of actin loops in muscle cell nuclei in the absence of nuclear α-actinin, suggesting Mlp84B has intrinsic actin cross-linking activity, which may complement α-actinin cross-linking activity at sites of actin filament anchorage. These results reveal a molecular mechanism for MLP stabilization of muscle and implicate reduced actin crosslinking as the primary destabilizing defect in MLP-associated cardiomyopathies. Our data support a model in which α-actinin and Mlp84B have important and overlapping functions at sites of actin filament anchorage to preserve muscle structure and function.
Collapse
Affiliation(s)
- Kathleen A. Clark
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
- Department of Biology, University of Utah, Salt Lake City, UT 84112
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112
| | - Julie L. Kadrmas
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
43
|
Chung CS, Hutchinson KR, Methawasin M, Saripalli C, Smith JE, Hidalgo CG, Luo X, Labeit S, Guo C, Granzier HL. Shortening of the elastic tandem immunoglobulin segment of titin leads to diastolic dysfunction. Circulation 2013; 128:19-28. [PMID: 23709671 DOI: 10.1161/circulationaha.112.001268] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Diastolic dysfunction is a poorly understood but clinically pervasive syndrome that is characterized by increased diastolic stiffness. Titin is the main determinant of cellular passive stiffness. However, the physiological role that the tandem immunoglobulin (Ig) segment of titin plays in stiffness generation and whether shortening this segment is sufficient to cause diastolic dysfunction need to be established. METHODS AND RESULTS We generated a mouse model in which 9 Ig-like domains (Ig3-Ig11) were deleted from the proximal tandem Ig segment of the spring region of titin (IG KO). Exon microarray analysis revealed no adaptations in titin splicing, whereas novel phospho-specific antibodies did not detect changes in titin phosphorylation. Passive myocyte stiffness was increased in the IG KO, and immunoelectron microscopy revealed increased extension of the remaining titin spring segments as the sole likely underlying mechanism. Diastolic stiffness was increased at the tissue and organ levels, with no consistent changes in extracellular matrix composition or extracellular matrix-based passive stiffness, supporting a titin-based mechanism for in vivo diastolic dysfunction. Additionally, IG KO mice have a reduced exercise tolerance, a phenotype often associated with diastolic dysfunction. CONCLUSIONS Increased titin-based passive stiffness is sufficient to cause diastolic dysfunction with exercise intolerance.
Collapse
Affiliation(s)
- Charles S Chung
- Department of Physiology, University of Arizona, PO Box245051, Tucson AZ 85724, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Biophysical Forces Modulate the Costamere and Z-Disc for Sarcomere Remodeling in Heart Failure. BIOPHYSICS OF THE FAILING HEART 2013. [DOI: 10.1007/978-1-4614-7678-8_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
Abstract
Current concepts of mechanosensation are general and applicable to almost every cell type. However, striated muscle cells are distinguished by their ability to generate strong forces via actin/myosin interaction, and this process is fine-tuned for optimum contractility. This aspect, unique for actively contracting cells, may be defined as "sensing of the magnitude and dynamics of contractility," as opposed to the well-known concepts of the "perception of extracellular mechanical stimuli." The acto/myosin interaction, by producing changes in ATP, ADP, Pi, and force on a millisecond timescale, may be regarded as a novel and previously unappreciated mechanosensory mechanism. In addition, sarcomeric mechanosensory structures, such as the Z-disc, are directly linked to autophagy, survival, and cell death-related pathways. One emerging example is telethonin and its ability to interfere with p53 metabolism and hence apoptosis (mechanoptosis). In this article, we introduce contractility per se as an important mechanosensory mechanism, and we differentiate extracellular from intracellular mechanosensory effects.
Collapse
Affiliation(s)
- Ralph Knöll
- Heart Science Section, National Heart & Lung Institute, Imperial College, London W12 0NN, UK.
| | | |
Collapse
|
46
|
Formation, contraction, and mechanotransduction of myofribrils in cardiac development: clues from genetics. Biochem Res Int 2012; 2012:504906. [PMID: 22720160 PMCID: PMC3376475 DOI: 10.1155/2012/504906] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 04/11/2012] [Accepted: 04/15/2012] [Indexed: 01/24/2023] Open
Abstract
Congenital heart disease (CHD) is the most common birth defect in humans. It is a leading infant mortality factor worldwide, caused by defective cardiac development. Mutations in transcription factors, signalling and structural molecules have been shown to contribute to the genetic component of CHD. Recently, mutations in genes encoding myofibrillar proteins expressed in the embryonic heart have also emerged as an important genetic causative factor of the disease, which implies that the contraction of the early heart primordium contributes to its morphogenesis. This notion is supported by increasing evidence suggesting that not only contraction but also formation, mechanosensing, and mechanotransduction of the cardiac myofibrillar proteins influence heart development. In this paper, we summarize the genetic clues supporting this idea.
Collapse
|
47
|
The diversification of the LIM superclass at the base of the metazoa increased subcellular complexity and promoted multicellular specialization. PLoS One 2012; 7:e33261. [PMID: 22438907 PMCID: PMC3305314 DOI: 10.1371/journal.pone.0033261] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 02/07/2012] [Indexed: 01/15/2023] Open
Abstract
Background Throughout evolution, the LIM domain has been deployed in many different domain configurations, which has led to the formation of a large and distinct group of proteins. LIM proteins are involved in relaying stimuli received at the cell surface to the nucleus in order to regulate cell structure, motility, and division. Despite their fundamental roles in cellular processes and human disease, little is known about the evolution of the LIM superclass. Results We have identified and characterized all known LIM domain-containing proteins in six metazoans and three non-metazoans. In addition, we performed a phylogenetic analysis on all LIM domains and, in the process, have identified a number of novel non-LIM domains and motifs in each of these proteins. Based on these results, we have formalized a classification system for LIM proteins, provided reasonable timing for class and family origin events; and identified lineage-specific loss events. Our analysis is the first detailed description of the full set of LIM proteins from the non-bilaterian species examined in this study. Conclusion Six of the 14 LIM classes originated in the stem lineage of the Metazoa. The expansion of the LIM superclass at the base of the Metazoa undoubtedly contributed to the increase in subcellular complexity required for the transition from a unicellular to multicellular lifestyle and, as such, was a critically important event in the history of animal multicellularity.
Collapse
|
48
|
Li A, Ponten F, dos Remedios CG. The interactome of LIM domain proteins: The contributions of LIM domain proteins to heart failure and heart development. Proteomics 2012; 12:203-25. [DOI: 10.1002/pmic.201100492] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 11/07/2011] [Accepted: 11/08/2011] [Indexed: 12/22/2022]
|
49
|
The sarcomeric Z-disc and Z-discopathies. J Biomed Biotechnol 2011; 2011:569628. [PMID: 22028589 PMCID: PMC3199094 DOI: 10.1155/2011/569628] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 08/12/2011] [Indexed: 02/06/2023] Open
Abstract
The sarcomeric Z-disc defines the lateral borders of the sarcomere and has primarily been seen as a structure important for mechanical stability. This view has changed dramatically within the last one or two decades. A multitude of novel Z-disc proteins and their interacting partners have been identified, which has led to the identification of additional functions and which have now been assigned to this structure. This includes its importance for intracellular signalling, for mechanosensation and mechanotransduction in particular, an emerging importance for protein turnover and autophagy, as well as its molecular links to the t-tubular system and the sarcoplasmic reticulum. Moreover, the discovery of mutations in a wide variety of Z-disc proteins, which lead to perturbations of several of the above-mentioned systems, gives rise to a diverse group of diseases which can be termed Z-discopathies. This paper provides a brief overview of these novel aspects as well as points to future research directions.
Collapse
|
50
|
de Tombe PP, Granzier HL. The cytoskeleton and the cellular transduction of mechanical strain in the heart: a special issue. Pflugers Arch 2011; 462:1-2. [PMID: 21594569 DOI: 10.1007/s00424-011-0976-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 05/05/2011] [Accepted: 05/06/2011] [Indexed: 01/15/2023]
|