1
|
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
|
2
|
Qi T, Zhang J, Zhang K, Zhang W, Song Y, Lian K, Kan C, Han F, Hou N, Sun X. Unraveling the role of the FHL family in cardiac diseases: Mechanisms, implications, and future directions. Biochem Biophys Res Commun 2024; 694:149468. [PMID: 38183876 DOI: 10.1016/j.bbrc.2024.149468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
Heart diseases are a major cause of morbidity and mortality worldwide. Understanding the molecular mechanisms underlying these diseases is essential for the development of effective diagnostic and therapeutic strategies. The FHL family consists of five members: FHL1, FHL2, FHL3, FHL4, and FHL5/Act. These members exhibit different expression patterns in various tissues including the heart. FHL family proteins are implicated in cardiac remodeling, regulation of metabolic enzymes, and cardiac biomechanical stress perception. A large number of studies have explored the link between FHL family proteins and cardiac disease, skeletal muscle disease, and ovarian metabolism, but a comprehensive and in-depth understanding of the specific molecular mechanisms targeting FHL on cardiac disease is lacking. The aim of this review is to explore the structure and function of FHL family members, to comprehensively elucidate the mechanisms by which they regulate the heart, and to explore in depth the changes in FHL family members observed in different cardiac disorders, as well as the effects of mutations in FHL proteins on heart health.
Collapse
Affiliation(s)
- Tongbing Qi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Wenqiang Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Yixin Song
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Kexin Lian
- Department of Nephrology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| |
Collapse
|
3
|
Elzamzami FD, Samal A, Arun AS, Dharmaraj T, Prasad NR, Rendon-Jonguitud A, DeVine L, Walston JD, Cole RN, Wilson KL. Native lamin A/C proteomes and novel partners from heart and skeletal muscle in a mouse chronic inflammation model of human frailty. Front Cell Dev Biol 2023; 11:1240285. [PMID: 37936983 PMCID: PMC10626543 DOI: 10.3389/fcell.2023.1240285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/05/2023] [Indexed: 11/09/2023] Open
Abstract
Clinical frailty affects ∼10% of people over age 65 and is studied in a chronically inflamed (Interleukin-10 knockout; "IL10-KO") mouse model. Frailty phenotypes overlap the spectrum of diseases ("laminopathies") caused by mutations in LMNA. LMNA encodes nuclear intermediate filament proteins lamin A and lamin C ("lamin A/C"), important for tissue-specific signaling, metabolism and chromatin regulation. We hypothesized that wildtype lamin A/C associations with tissue-specific partners are perturbed by chronic inflammation, potentially contributing to dysfunction in frailty. To test this idea we immunoprecipitated native lamin A/C and associated proteins from skeletal muscle, hearts and brains of old (21-22 months) IL10-KO versus control C57Bl/6 female mice, and labeled with Tandem Mass Tags for identification and quantitation by mass spectrometry. We identified 502 candidate lamin-binding proteins from skeletal muscle, and 340 from heart, including 62 proteins identified in both tissues. Candidates included frailty phenotype-relevant proteins Perm1 and Fam210a, and nuclear membrane protein Tmem38a, required for muscle-specific genome organization. These and most other candidates were unaffected by IL10-KO, but still important as potential lamin A/C-binding proteins in native heart or muscle. A subset of candidates (21 in skeletal muscle, 30 in heart) showed significantly different lamin A/C-association in an IL10-KO tissue (p < 0.05), including AldoA and Gins3 affected in heart, and Lmcd1 and Fabp4 affected in skeletal muscle. To screen for binding, eleven candidates plus prelamin A and emerin controls were arrayed as synthetic 20-mer peptides (7-residue stagger) and incubated with recombinant purified lamin A "tail" residues 385-646 under relatively stringent conditions. We detected strong lamin A binding to peptides solvent exposed in Lmcd1, AldoA, Perm1, and Tmem38a, and plausible binding to Csrp3 (muscle LIM protein). These results validated both proteomes as sources for native lamin A/C-binding proteins in heart and muscle, identified four candidate genes for Emery-Dreifuss muscular dystrophy (CSRP3, LMCD1, ALDOA, and PERM1), support a lamin A-interactive molecular role for Tmem38A, and supported the hypothesis that lamin A/C interactions with at least two partners (AldoA in heart, transcription factor Lmcd1 in muscle) are altered in the IL10-KO model of frailty.
Collapse
Affiliation(s)
- Fatima D. Elzamzami
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Arushi Samal
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Adith S. Arun
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Tejas Dharmaraj
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Neeti R. Prasad
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alex Rendon-Jonguitud
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lauren DeVine
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jeremy D. Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Robert N. Cole
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Katherine L. Wilson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
Park J, Bird JE. The actin cytoskeleton in hair bundle development and hearing loss. Hear Res 2023; 436:108817. [PMID: 37300948 PMCID: PMC10408727 DOI: 10.1016/j.heares.2023.108817] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023]
Abstract
Inner ear hair cells assemble mechanosensitive hair bundles on their apical surface that transduce sounds and accelerations. Each hair bundle is comprised of ∼ 100 individual stereocilia that are arranged into rows of increasing height and width; their specific and precise architecture being necessary for mechanoelectrical transduction (MET). The actin cytoskeleton is fundamental to establishing this architecture, not only by forming the structural scaffold shaping each stereocilium, but also by composing rootlets and the cuticular plate that together provide a stable foundation supporting each stereocilium. In concert with the actin cytoskeleton, a large assortment of actin-binding proteins (ABPs) function to cross-link actin filaments into specific topologies, as well as control actin filament growth, severing, and capping. These processes are individually critical for sensory transduction and are all disrupted in hereditary forms of human hearing loss. In this review, we provide an overview of actin-based structures in the hair bundle and the molecules contributing to their assembly and functional properties. We also highlight recent advances in mechanisms driving stereocilia elongation and how these processes are tuned by MET.
Collapse
Affiliation(s)
- Jinho Park
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, United States; Myology Institute, University of Florida, Gainesville, FL 32610, United States
| | - Jonathan E Bird
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, United States; Myology Institute, University of Florida, Gainesville, FL 32610, United States.
| |
Collapse
|
5
|
Mgrditchian T, Brown-Clay J, Hoffmann C, Müller T, Filali L, Ockfen E, Mao X, Moreau F, Casellas CP, Kaoma T, Mittelbronn M, Thomas C. Actin cytoskeleton depolymerization increases matrix metalloproteinase gene expression in breast cancer cells by promoting translocation of cysteine-rich protein 2 to the nucleus. Front Cell Dev Biol 2023; 11:1100938. [PMID: 37266453 PMCID: PMC10229898 DOI: 10.3389/fcell.2023.1100938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 04/21/2023] [Indexed: 06/03/2023] Open
Abstract
The actin cytoskeleton plays a critical role in cancer cell invasion and metastasis; however, the coordination of its multiple functions remains unclear. Actin dynamics in the cytoplasm control the formation of invadopodia, which are membrane protrusions that facilitate cancer cell invasion by focusing the secretion of extracellular matrix-degrading enzymes, including matrix metalloproteinases (MMPs). In this study, we investigated the nuclear role of cysteine-rich protein 2 (CRP2), a two LIM domain-containing F-actin-binding protein that we previously identified as a cytoskeletal component of invadopodia, in breast cancer cells. We found that F-actin depolymerization stimulates the translocation of CRP2 into the nucleus, resulting in an increase in the transcript levels of pro-invasive and pro-metastatic genes, including several members of the MMP gene family. We demonstrate that in the nucleus, CRP2 interacts with the transcription factor serum response factor (SRF), which is crucial for the expression of MMP-9 and MMP-13. Our data suggest that CRP2 and SRF cooperate to modulate of MMP expression levels. Furthermore, Kaplan-Meier analysis revealed a significant association between high-level expression of SRF and shorter overall survival and distant metastasis-free survival in breast cancer patients with a high CRP2 expression profile. Our findings suggest a model in which CRP2 mediates the coordination of cytoplasmic and nuclear processes driven by actin dynamics, ultimately resulting in the induction of invasive and metastatic behavior in breast cancer cells.
Collapse
Affiliation(s)
- Takouhie Mgrditchian
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Joshua Brown-Clay
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Céline Hoffmann
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Tanja Müller
- Department of Cancer Research, Luxembourg Centre of Neuropathology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Liza Filali
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Elena Ockfen
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Xianqing Mao
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Flora Moreau
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Carla Pou Casellas
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Tony Kaoma
- Bioinformatics Platform, Luxembourg, Luxembourg
| | - Michel Mittelbronn
- Department of Cancer Research, Luxembourg Centre of Neuropathology, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-surAlzette, Luxembourg
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-surAlzette, Luxembourg
- Department of Life Science and Medicine (DLSM), University of Luxembourg, Esch-surAlzette, Luxembourg
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
| | - Clément Thomas
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
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
|
Wishard R, Jayaram M, Ramesh SR, Nongthomba U. Spatial and temporal requirement of Mlp60A isoforms during muscle development and function in Drosophila melanogaster. Exp Cell Res 2023; 422:113430. [PMID: 36423661 DOI: 10.1016/j.yexcr.2022.113430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022]
Abstract
Many myofibrillar proteins undergo isoform switching in a spatio-temporal manner during muscle development. The biological significance of the variants of several of these myofibrillar proteins remains elusive. One such myofibrillar protein, the Muscle LIM Protein (MLP), is a vital component of the Z-discs. In this paper, we show that one of the Drosophila MLP encoding genes, Mlp60A, gives rise to two isoforms: a short (279 bp, 10 kDa) and a long (1461 bp, 54 kDa) one. The short isoform is expressed throughout development, but the long isoform is adult-specific, being the dominant of the two isoforms in the indirect flight muscles (IFMs). A concomitant, muscle-specific knockdown of both isoforms leads to partial developmental lethality, with most of the surviving flies being flight defective. A global loss of both isoforms in a Mlp60A-null background also leads to developmental lethality, with muscle defects in the individuals that survive to the third instar larval stage. This lethality could be rescued partially by a muscle-specific overexpression of the short isoform. Genetic perturbation of only the long isoform, through a P-element insertion in the long isoform-specific coding sequence, leads to defective flight, in around 90% of the flies. This phenotype was completely rescued when the P-element insertion was precisely excised from the locus. Hence, our data show that the two Mlp60A isoforms are functionally specialized: the short isoform being essential for normal embryonic muscle development and the long isoform being necessary for normal adult flight muscle function.
Collapse
Affiliation(s)
- Rohan Wishard
- Department of Molecular Reproduction, Development and Genetics; Indian Institute of Science, Bengaluru, 560012, India.
| | - Mohan Jayaram
- Department of Molecular Reproduction, Development and Genetics; Indian Institute of Science, Bengaluru, 560012, India; Department of Studies in Zoology, University of Mysore, Manasgangotri, Mysuru, 570006, India
| | - Saraf R Ramesh
- Department of Studies in Zoology, University of Mysore, Manasgangotri, Mysuru, 570006, India; Department of Life Sciences, Pooja Bhagvat Memorial Mahajana Education Center, K. R. S. Road, Mysuru, 570016, India
| | - Upendra Nongthomba
- Department of Molecular Reproduction, Development and Genetics; Indian Institute of Science, Bengaluru, 560012, India.
| |
Collapse
|
8
|
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
|
9
|
Riaz M, Park J, Sewanan LR, Ren Y, Schwan J, Das SK, Pomianowski PT, Huang Y, Ellis MW, Luo J, Liu J, Song L, Chen IP, Qiu C, Yazawa M, Tellides G, Hwa J, Young LH, Yang L, Marboe CC, Jacoby DL, Campbell SG, Qyang Y. Muscle LIM Protein Force-Sensing Mediates Sarcomeric Biomechanical Signaling in Human Familial Hypertrophic Cardiomyopathy. Circulation 2022; 145:1238-1253. [PMID: 35384713 PMCID: PMC9109819 DOI: 10.1161/circulationaha.121.056265] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Familial hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac disease and is typically caused by mutations in genes encoding sarcomeric proteins that regulate cardiac contractility. HCM manifestations include left ventricular hypertrophy and heart failure, arrythmias, and sudden cardiac death. How dysregulated sarcomeric force production is sensed and leads to pathological remodeling remains poorly understood in HCM, thereby inhibiting the efficient development of new therapeutics. METHODS Our discovery was based on insights from a severe phenotype of an individual with HCM and a second genetic alteration in a sarcomeric mechanosensing protein. We derived cardiomyocytes from patient-specific induced pluripotent stem cells and developed robust engineered heart tissues by seeding induced pluripotent stem cell-derived cardiomyocytes into a laser-cut scaffold possessing native cardiac fiber alignment to study human cardiac mechanobiology at both the cellular and tissue levels. Coupled with computational modeling for muscle contraction and rescue of disease phenotype by gene editing and pharmacological interventions, we have identified a new mechanotransduction pathway in HCM, shown to be essential in modulating the phenotypic expression of HCM in 5 families bearing distinct sarcomeric mutations. RESULTS Enhanced actomyosin crossbridge formation caused by sarcomeric mutations in cardiac myosin heavy chain (MYH7) led to increased force generation, which, when coupled with slower twitch relaxation, destabilized the MLP (muscle LIM protein) stretch-sensing complex at the Z-disc. Subsequent reduction in the sarcomeric muscle LIM protein level caused disinhibition of calcineurin-nuclear factor of activated T-cells signaling, which promoted cardiac hypertrophy. We demonstrate that the common muscle LIM protein-W4R variant is an important modifier, exacerbating the phenotypic expression of HCM, but alone may not be a disease-causing mutation. By mitigating enhanced actomyosin crossbridge formation through either genetic or pharmacological means, we alleviated stress at the Z-disc, preventing the development of hypertrophy associated with sarcomeric mutations. CONCLUSIONS Our studies have uncovered a novel biomechanical mechanism through which dysregulated sarcomeric force production is sensed and leads to pathological signaling, remodeling, and hypertrophic responses. Together, these establish the foundation for developing innovative mechanism-based treatments for HCM that stabilize the Z-disc MLP-mechanosensory complex.
Collapse
Affiliation(s)
- Muhammad Riaz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Jinkyu Park
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Lorenzo R. Sewanan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Yongming Ren
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Jonas Schwan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Subhash K. Das
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | | | - Yan Huang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Matthew W. Ellis
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Jiesi Luo
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Juli Liu
- Department of Pediatrics, Anatomy and Cell Biology, Indiana University, Indianapolis, IN, USA
| | - Loujin Song
- Department of Rehabilitation and Regenerative Medicine, Columbia Stem Cell Initiative, Columbia University, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
| | - I-Ping Chen
- Department of Oral Health and Diagnostic Sciences, University of Connecticut Health, Farmington, CT, USA
| | | | - Masayuki Yazawa
- Department of Rehabilitation and Regenerative Medicine, Columbia Stem Cell Initiative, Columbia University, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
| | | | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Lawrence H. Young
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Lei Yang
- Department of Pediatrics, Anatomy and Cell Biology, Indiana University, Indianapolis, IN, USA
| | - Charles C. Marboe
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Daniel L. Jacoby
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Stuart G. Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Yibing Qyang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
10
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
11
|
Langer HT, Mossakowski AA, Avey AM, Wohlgemuth RP, Smith LR, Zbinden-Foncea H, Baar K. A mutation in desmin makes skeletal muscle less vulnerable to acute muscle damage after eccentric loading in rats. FASEB J 2021; 35:e21860. [PMID: 34411340 PMCID: PMC9292853 DOI: 10.1096/fj.202100711rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/15/2021] [Accepted: 08/02/2021] [Indexed: 01/23/2023]
Abstract
Desminopathy is the most common intermediate filament disease in humans. The most frequent mutation causing desminopathy in patients is a R350P DES missense mutation. We have developed a rat model with an analogous mutation in R349P Des. To investigate the role of R349P Des in mechanical loading, we stimulated the sciatic nerve of wild‐type littermates (WT) (n = 6) and animals carrying the mutation (MUT) (n = 6) causing a lengthening contraction of the dorsi flexor muscles. MUT animals showed signs of ongoing regeneration at baseline as indicated by a higher number of central nuclei (genotype: P < .0001). While stimulation did not impact central nuclei, we found an increased number of IgG positive fibers (membrane damage indicator) after eccentric contractions with both genotypes (stimulation: P < .01). Interestingly, WT animals displayed a more pronounced increase in IgG positive fibers with stimulation compared to MUT (interaction: P < .05). In addition to altered histology, molecular signaling on the protein level differed between WT and MUT. The membrane repair protein dysferlin decreased with eccentric loading in WT but increased in MUT (interaction: P < .05). The autophagic substrate p62 was increased in both genotypes with loading (stimulation: P < .05) but tended to be more elevated in WT (interaction: P = .05). Caspase 3 levels, a central regulator of apoptotic cell death, was increased with stimulation in both genotypes (stimulation: P < .01) but more so in WT animals (interaction: P < .0001). Overall, our data indicate that R349P Des rats have a lower susceptibility to structural muscle damage of the cytoskeleton and sarcolemma with acute eccentric loading.
Collapse
Affiliation(s)
- Henning T Langer
- Functional Molecular Biology Laboratory, Department of Physiology and Membrane Biology, University of California, Davis, California, USA
| | - Agata A Mossakowski
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alec M Avey
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA
| | - Ross P Wohlgemuth
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA
| | - Lucas R Smith
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA
| | - Herman Zbinden-Foncea
- Exercise Physiology Laboratory, School of Kinesiology, Universidad Finis Terrae, Santiago, Chile
| | - Keith Baar
- Functional Molecular Biology Laboratory, Department of Physiology and Membrane Biology, University of California, Davis, California, USA.,Neurobiology, Physiology and Behavior, University of California, Davis, California, USA.,VA Northern California Health Care System, Mather, California, USA
| |
Collapse
|
12
|
Anderson CA, Kovar DR, Gardel ML, Winkelman JD. LIM domain proteins in cell mechanobiology. Cytoskeleton (Hoboken) 2021; 78:303-311. [PMID: 34028199 DOI: 10.1002/cm.21677] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022]
Abstract
The actin cytoskeleton is important for maintaining mechanical homeostasis in adherent cells, largely through its regulation of adhesion and cortical tension. The LIM (Lin-11, Isl1, MEC-3) domain-containing proteins are involved in a myriad of cellular mechanosensitive pathways. Recent work has discovered that LIM domains bind to mechanically stressed actin filaments, suggesting a novel and widely conserved mechanism of mechanosensing. This review summarizes the current state of knowledge of LIM protein mechanosensitivity.
Collapse
Affiliation(s)
- Caitlin A Anderson
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, USA
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, USA.,Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA
| | - Margaret L Gardel
- Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois, USA.,James Franck Institute, University of Chicago, Chicago, Illinois, USA.,Department of Physics, University of Chicago, Chicago, Illinois, USA.,Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA
| | - Jonathan D Winkelman
- Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
13
|
The multiple roles of actin-binding proteins at invadopodia. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021. [PMID: 33962752 DOI: 10.1016/bs.ircmb.2021.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Invadopodia are actin-rich membrane protrusions that facilitate cancer cell dissemination by focusing on proteolytic activity and clearing paths for migration through physical barriers, such as basement membranes, dense extracellular matrices, and endothelial cell junctions. Invadopodium formation and activity require spatially and temporally regulated changes in actin filament organization and dynamics. About three decades of research have led to a remarkable understanding of how these changes are orchestrated by sequential recruitment and coordinated activity of different sets of actin-binding proteins. In this chapter, we provide an update on the roles of the actin cytoskeleton during the main stages of invadopodium development with a particular focus on actin polymerization machineries and production of pushing forces driving extracellular matrix remodeling.
Collapse
|
14
|
Abstract
Age-associated changes in gene expression in skeletal muscle of healthy individuals reflect accumulation of damage and compensatory adaptations to preserve tissue integrity. To characterize these changes, RNA was extracted and sequenced from muscle biopsies collected from 53 healthy individuals (22-83 years old) of the GESTALT study of the National Institute on Aging-NIH. Expression levels of 57,205 protein-coding and non-coding RNAs were studied as a function of aging by linear and negative binomial regression models. From both models, 1134 RNAs changed significantly with age. The most differentially abundant mRNAs encoded proteins implicated in several age-related processes, including cellular senescence, insulin signaling, and myogenesis. Specific mRNA isoforms that changed significantly with age in skeletal muscle were enriched for proteins involved in oxidative phosphorylation and adipogenesis. Our study establishes a detailed framework of the global transcriptome and mRNA isoforms that govern muscle damage and homeostasis with age.
Collapse
|
15
|
Higaki T, Akita K, Katoh K. Coefficient of variation as an image-intensity metric for cytoskeleton bundling. Sci Rep 2020; 10:22187. [PMID: 33349642 PMCID: PMC7752905 DOI: 10.1038/s41598-020-79136-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/27/2020] [Indexed: 12/17/2022] Open
Abstract
The evaluation of cytoskeletal bundling is a fundamental experimental method in the field of cell biology. Although the skewness of the pixel intensity distribution derived from fluorescently-labeled cytoskeletons has been widely used as a metric to evaluate the degree of bundling in digital microscopy images, its versatility has not been fully validated. Here, we applied the coefficient of variation (CV) of intensity values as an alternative metric, and compared its performance with skewness. In synthetic images representing extremely bundled conditions, the CV successfully detected degrees of bundling that could not be distinguished by skewness. On actual microscopy images, CV was better than skewness, especially on variable-angle epifluorescence microscopic images or stimulated emission depletion and confocal microscopy images of very small areas of around 1 μm2. When blur or noise was added to synthetic images, CV was found to be robust to blur but deleteriously affected by noise, whereas skewness was robust to noise but deleteriously affected by blur. For confocal images, CV and skewness showed similar sensitivity to noise, possibly because optical blurring is often present in microscopy images. Therefore, in practical use with actual microscopy images, CV may be more appropriate than skewness, unless the image is extremely noisy.
Collapse
Affiliation(s)
- Takumi Higaki
- International Research Organization for Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, Japan.
| | - Kae Akita
- Department of Chemical Biological Science, Faculty of Science, Japan Women's University, 2-8-1 Mejirodai, Bunkyo-ku, Tokyo, Japan
| | - Kaoru Katoh
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| |
Collapse
|
16
|
Olie CS, van der Wal E, Cikes D, Maton L, de Greef JC, Lin IH, Chen YF, Kareem E, Penninger JM, Kessler BM, Raz V. Cytoskeletal disorganization underlies PABPN1-mediated myogenic disability. Sci Rep 2020; 10:17621. [PMID: 33077830 PMCID: PMC7572364 DOI: 10.1038/s41598-020-74676-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 09/28/2020] [Indexed: 12/30/2022] Open
Abstract
Muscle wasting and atrophy are regulated by multiple molecular processes, including mRNA processing. Reduced levels of the polyadenylation binding protein nucleus 1 (PABPN1), a multifactorial regulator of mRNA processing, cause muscle atrophy. A proteomic study in muscles with reduced PABPN1 levels suggested dysregulation of sarcomeric and cytoskeletal proteins. Here we investigated the hypothesis that reduced PABPN1 levels lead to an aberrant organization of the cytoskeleton. MURC, a plasma membrane-associated protein, was found to be more abundant in muscles with reduced PABPN1 levels, and it was found to be expressed at regions showing regeneration. A polarized cytoskeletal organization is typical for muscle cells, but muscle cells with reduced PABPN1 levels (named as shPAB) were characterized by a disorganized cytoskeleton that lacked polarization. Moreover, cell mechanical features and myogenic differentiation were significantly reduced in shPAB cells. Importantly, restoring cytoskeletal stability, by actin overexpression, was beneficial for myogenesis, expression of sarcomeric proteins and proper localization of MURC in shPAB cell cultures and in shPAB muscle bundle. We suggest that poor cytoskeletal mechanical features are caused by altered expression levels of cytoskeletal proteins and contribute to muscle wasting and atrophy.
Collapse
Affiliation(s)
| | - Erik van der Wal
- Human Genetics Department, Leiden University Medical Center, Leiden, The Netherlands
| | - Domagoj Cikes
- IMBA-Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Loes Maton
- Human Genetics Department, Leiden University Medical Center, Leiden, The Netherlands
| | - Jessica C de Greef
- Human Genetics Department, Leiden University Medical Center, Leiden, The Netherlands
| | - I-Hsuan Lin
- VYM Genome Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Fan Chen
- College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Elsayad Kareem
- Advanced Microscopy Facility, Vienna Biocenter Core Facilities, Vienna Biocenter (VBC), Vienna, Austria
| | - Josef M Penninger
- IMBA-Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Benedikt M Kessler
- Target Discovery Institute, Nuffield, Department of Medicine, University of Oxford, Oxford, UK
| | - Vered Raz
- Human Genetics Department, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
17
|
Huang X, Qu R, Ouyang J, Zhong S, Dai J. An Overview of the Cytoskeleton-Associated Role of PDLIM5. Front Physiol 2020; 11:975. [PMID: 32848888 PMCID: PMC7426503 DOI: 10.3389/fphys.2020.00975] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/16/2020] [Indexed: 01/08/2023] Open
Abstract
Regenerative medicine represented by stem cell technology has become one of the pillar medical technologies for human disease treatment. Cytoskeleton plays important roles in maintaining cell morphology, bearing external forces, and maintaining the effectiveness of cell internal structure, among which cytoskeleton related proteins are involved in and play an indispensable role in the changes of cytoskeleton. PDLIM5 is a cytoskeleton-related protein that, like other cytoskeletal proteins, acts as a binding protein. PDZ and LIM domain 5 (PDLIM5), also known as ENH (Enigma homolog), is a cytoplasmic protein with a molecular mass of about 63 KDa that consists of a PDZ domain at the N-terminus and three LIM domains at the C-terminus. PDLIM5 binds to the cytoskeleton and membrane proteins through its PDZ domain and interacts with various signaling molecules, including protein kinases and transcription factors, through its LIM domain. As a cytoskeleton-related protein, PDLIM5 plays an important role in regulating cell proliferation, differentiation and cell fate decision in multiple tissues and cell types. In this review, we briefly summarize the state of knowledge on the PDLIM5 gene, structural properties, and molecular functional mechanisms of the PDLIM5 protein, and its role in cells, tissues, and organ systems, and describe the possible underlying molecular signaling pathways. In the last part of this review, we will focus on discussing the limitations of existing research and the future prospects of PDLIM5 research in turn.
Collapse
Affiliation(s)
- Xiaolan Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shizhen Zhong
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
18
|
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
|
19
|
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: 6] [Impact Index Per Article: 1.5] [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
|
20
|
Ward M, Iskratsch T. Mix and (mis-)match - The mechanosensing machinery in the changing environment of the developing, healthy adult and diseased heart. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118436. [PMID: 30742931 PMCID: PMC7042712 DOI: 10.1016/j.bbamcr.2019.01.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/07/2019] [Accepted: 01/29/2019] [Indexed: 01/01/2023]
Abstract
The composition and the stiffness of cardiac microenvironment change during development and/or in heart disease. Cardiomyocytes (CMs) and their progenitors sense these changes, which decides over the cell fate and can trigger CM (progenitor) proliferation, differentiation, de-differentiation or death. The field of mechanobiology has seen a constant increase in output that also includes a wealth of new studies specific to cardiac or cardiomyocyte mechanosensing. As a result, mechanosensing and transduction in the heart is increasingly being recognised as a main driver of regulating the heart formation and function. Recent work has for instance focused on measuring the molecular, physical and mechanical changes of the cellular environment - as well as intracellular contributors to the passive stiffness of the heart. On the other hand, a variety of new studies shed light into the molecular machinery that allow the cardiomyocytes to sense these properties. Here we want to discuss the recent work on this topic, but also specifically focus on how the different components are regulated at various stages during development, in health or disease in order to highlight changes that might contribute to disease progression and heart failure.
Collapse
Key Words
- cm, cardiomyocytes
- hcm, hypertrophic cardiomyopathy
- dcm, dilated cardiomyopathy
- icm, idiopathic cardiomyopathy
- myh, myosin heavy chain
- tnnt, troponin t
- tnni, troponin i
- afm, atomic force microscope
- mre, magnetic resonance elastography
- swe, ultrasound cardiac shear-wave elastography
- lv, left ventricle
- lox, lysyl oxidase
- loxl, lysyl oxidase like protein
- lh, lysyl hydroxylase
- lys, lysin
- lccs, lysald-derived collagen crosslinks
- hlccs, hylald-derived collagen crosslinks
- pka, protein kinase a
- pkc, protein kinase c
- vash1, vasohibin-1
- svbp, small vasohibin binding protein
- tcp, tubulin carboxypeptidase
- ttl, tubulin tyrosine ligase
- mrtf, myocardin-related transcription factor
- gap, gtpase activating protein
- gef, guanine nucleotide exchange factor
Collapse
Affiliation(s)
- Matthew Ward
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom.
| |
Collapse
|
21
|
Mukund K, Subramaniam S. Skeletal muscle: A review of molecular structure and function, in health and disease. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1462. [PMID: 31407867 PMCID: PMC6916202 DOI: 10.1002/wsbm.1462] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/03/2019] [Accepted: 07/03/2019] [Indexed: 12/11/2022]
Abstract
Decades of research in skeletal muscle physiology have provided multiscale insights into the structural and functional complexity of this important anatomical tissue, designed to accomplish the task of generating contraction, force and movement. Skeletal muscle can be viewed as a biomechanical device with various interacting components including the autonomic nerves for impulse transmission, vasculature for efficient oxygenation, and embedded regulatory and metabolic machinery for maintaining cellular homeostasis. The "omics" revolution has propelled a new era in muscle research, allowing us to discern minute details of molecular cross-talk required for effective coordination between the myriad interacting components for efficient muscle function. The objective of this review is to provide a systems-level, comprehensive mapping the molecular mechanisms underlying skeletal muscle structure and function, in health and disease. We begin this review with a focus on molecular mechanisms underlying muscle tissue development (myogenesis), with an emphasis on satellite cells and muscle regeneration. We next review the molecular structure and mechanisms underlying the many structural components of the muscle: neuromuscular junction, sarcomere, cytoskeleton, extracellular matrix, and vasculature surrounding muscle. We highlight aberrant molecular mechanisms and their possible clinical or pathophysiological relevance. We particularly emphasize the impact of environmental stressors (inflammation and oxidative stress) in contributing to muscle pathophysiology including atrophy, hypertrophy, and fibrosis. This article is categorized under: Physiology > Mammalian Physiology in Health and Disease Developmental Biology > Developmental Processes in Health and Disease Models of Systems Properties and Processes > Cellular Models.
Collapse
Affiliation(s)
- Kavitha Mukund
- Department of BioengineeringUniversity of CaliforniaSan DiegoCalifornia
| | - Shankar Subramaniam
- Department of Bioengineering, Bioinformatics & Systems BiologyUniversity of CaliforniaSan DiegoCalifornia
- Department of Computer Science and EngineeringUniversity of CaliforniaSan DiegoCalifornia
- Department of Cellular and Molecular Medicine and NanoengineeringUniversity of CaliforniaSan DiegoCalifornia
| |
Collapse
|
22
|
Li X, Lu WJ, Li Y, Wu F, Bai R, Ma S, Dong T, Zhang H, Lee AS, Wang Y, Lan F. MLP-deficient human pluripotent stem cell derived cardiomyocytes develop hypertrophic cardiomyopathy and heart failure phenotypes due to abnormal calcium handling. Cell Death Dis 2019; 10:610. [PMID: 31406109 PMCID: PMC6690906 DOI: 10.1038/s41419-019-1826-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/07/2019] [Accepted: 07/02/2019] [Indexed: 02/08/2023]
Abstract
Muscle LIM protein (MLP, CSRP3) is a key regulator of striated muscle function, and its mutations can lead to both hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) in patients. However, due to lack of human models, mechanisms underlining the pathogenesis of MLP defects remain unclear. In this study, we generated a knockout MLP/CSRP3 human embryonic stem cell (hESC) H9 cell line using CRISPR/Cas9 mediated gene disruption. CSRP3 disruption had no impact on the cardiac differentiation of H9 cells and led to confirmed MLP deficiency in hESC-derived cardiomyocytes (ESC-CMs). MLP-deficient hESC-CMs were found to develop phenotypic features of HCM early after differentiation, such as enlarged cell size, multinucleation, and disorganized sarcomeric ultrastructure. Cellular phenotypes of MLP-deficient hESC-CMs subsequently progressed to mimic heart failure (HF) by 30 days post differentiation, including exhibiting mitochondrial damage, increased ROS generation, and impaired Ca2+ handling. Pharmaceutical treatment with beta agonist, such as isoproterenol, was found to accelerate the manifestation of HCM and HF, consistent with transgenic animal models of MLP deficiency. Furthermore, restoration of Ca2+ homeostasis by verapamil prevented the development of HCM and HF phenotypes, suggesting that elevated intracellular Ca2+ concentration is a central mechanism for pathogenesis of MLP deficiency. In summary, MLP-deficient hESC-CMs recapitulate the pathogenesis of HCM and its progression toward HF, providing an important human model for investigation of CSRP3/MLP-associated disease pathogenesis. More importantly, correction of the autonomous dysfunction of Ca2+ handling was found to be an effective method for treating the in vitro development of cardiomyopathy disease phenotype.
Collapse
Affiliation(s)
- Xiaowei Li
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, 100029, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, 100029, Beijing, China
| | - Wen-Jing Lu
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, 100029, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, 100029, Beijing, China
| | - Ya'nan Li
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, 100029, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, 100029, Beijing, China
| | - Fujian Wu
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, 100029, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, 100029, Beijing, China
| | - Rui Bai
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, 100029, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, 100029, Beijing, China
| | - Shuhong Ma
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, 100029, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, 100029, Beijing, China
| | - Tao Dong
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, 100029, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, 100029, Beijing, China
| | - Hongjia Zhang
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, 100029, Beijing, China
| | - Andrew S Lee
- Center for Clinical Translation and Innovation, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| | - Yongming Wang
- The State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| | - Feng Lan
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, 100029, Beijing, China.
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, 100029, Beijing, China.
| |
Collapse
|
23
|
Terheyden-Keighley D, Fischer D. The role of muscle LIM protein in the nervous system. Neural Regen Res 2019; 14:1907-1908. [PMID: 31290445 PMCID: PMC6676870 DOI: 10.4103/1673-5374.259607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Daniel Terheyden-Keighley
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Germany
| | - Dietmar Fischer
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Germany
| |
Collapse
|
24
|
Capraro A, O'Meally D, Waters SA, Patel HR, Georges A, Waters PD. Waking the sleeping dragon: gene expression profiling reveals adaptive strategies of the hibernating reptile Pogona vitticeps. BMC Genomics 2019; 20:460. [PMID: 31170930 PMCID: PMC6555745 DOI: 10.1186/s12864-019-5750-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/29/2019] [Indexed: 12/30/2022] Open
Abstract
Background Hibernation is a physiological state exploited by many animals exposed to prolonged adverse environmental conditions associated with winter. Large changes in metabolism and cellular function occur, with many stress response pathways modulated to tolerate physiological challenges that might otherwise be lethal. Many studies have sought to elucidate the molecular mechanisms of mammalian hibernation, but detailed analyses are lacking in reptiles. Here we examine gene expression in the Australian central bearded dragon (Pogona vitticeps) using mRNA-seq and label-free quantitative mass spectrometry in matched brain, heart and skeletal muscle samples from animals at late hibernation, 2 days post-arousal and 2 months post-arousal. Results We identified differentially expressed genes in all tissues between hibernation and post-arousal time points; with 4264 differentially expressed genes in brain, 5340 differentially expressed genes in heart, and 5587 differentially expressed genes in skeletal muscle. Furthermore, we identified 2482 differentially expressed genes across all tissues. Proteomic analysis identified 743 proteins (58 differentially expressed) in brain, 535 (57 differentially expressed) in heart, and 337 (36 differentially expressed) in skeletal muscle. Tissue-specific analyses revealed enrichment of protective mechanisms in all tissues, including neuroprotective pathways in brain, cardiac hypertrophic processes in heart, and atrophy protective pathways in skeletal muscle. In all tissues stress response pathways were induced during hibernation, as well as evidence for gene expression regulation at transcription, translation and post-translation. Conclusions These results reveal critical stress response pathways and protective mechanisms that allow for maintenance of both tissue-specific function, and survival during hibernation in the central bearded dragon. Furthermore, we provide evidence for multiple levels of gene expression regulation during hibernation, particularly enrichment of miRNA-mediated translational repression machinery; a process that would allow for rapid and energy efficient reactivation of translation from mature mRNA molecules at arousal. This study is the first molecular investigation of its kind in a hibernating reptile, and identifies strategies not yet observed in other hibernators to cope stress associated with this remarkable state of metabolic depression. Electronic supplementary material The online version of this article (10.1186/s12864-019-5750-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexander Capraro
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, UNSW Sydney, Sydney, NSW, 2052, Australia.
| | - Denis O'Meally
- Institute for Applied Ecology, University of Canberra, Canberra, ACT, 2601, Australia.,Present address: Center for Gene Therapy, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Shafagh A Waters
- School of Women's & Children's Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Hardip R Patel
- John Curtin School of Medical Research, Australian National University, Canberra, 2601, ACT, Australia
| | - Arthur Georges
- Institute for Applied Ecology, University of Canberra, Canberra, ACT, 2601, Australia
| | - Paul D Waters
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, UNSW Sydney, Sydney, NSW, 2052, Australia
| |
Collapse
|
25
|
VanHecke GC, Abeywardana MY, Ahn YH. Proteomic Identification of Protein Glutathionylation in Cardiomyocytes. J Proteome Res 2019; 18:1806-1818. [PMID: 30831029 DOI: 10.1021/acs.jproteome.8b00986] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Reactive oxygen species (ROS) are important signaling molecules, but their overproduction is associated with many cardiovascular diseases, including cardiomyopathy. ROS induce various oxidative modifications, among which glutathionylation is one of the significant protein oxidations that occur under oxidative stress. Despite previous efforts, direct and site-specific identification of glutathionylated proteins in cardiomyocytes has been limited. In this report, we used a clickable glutathione approach in a HL-1 mouse cardiomyocyte cell line under exposure to hydrogen peroxide, finding 1763 glutathionylated peptides with specific Cys modification sites, which include many muscle-specific proteins. Bioinformatic and cluster analyses found 125 glutathionylated proteins, whose mutations or dysfunctions are associated with cardiomyopathy, many of which include sarcomeric structural and contractile proteins, chaperone, and other signaling or regulatory proteins. We further provide functional implication of glutathionylation for several identified proteins, including CSRP3/MLP and complex I, II, and III, by analyzing glutathionylated sites in their structures. Our report establishes a chemoselective method for direct identification of glutathionylated proteins and provides potential target proteins whose glutathionylation may contribute to muscle diseases.
Collapse
Affiliation(s)
- Garrett C VanHecke
- Department of Chemistry , Wayne State University , Detroit , Michigan 48202 , United States
| | | | - Young-Hoon Ahn
- Department of Chemistry , Wayne State University , Detroit , Michigan 48202 , United States
| |
Collapse
|
26
|
Hernandez-Carretero A, Weber N, LaBarge SA, Peterka V, Doan NYT, Schenk S, Osborn O. Cysteine- and glycine-rich protein 3 regulates glucose homeostasis in skeletal muscle. Am J Physiol Endocrinol Metab 2018; 315:E267-E278. [PMID: 29634311 PMCID: PMC6139493 DOI: 10.1152/ajpendo.00435.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Skeletal muscle is the major site of postprandial peripheral glucose uptake, but in obesity-induced insulin-resistant states insulin-stimulated glucose disposal is markedly impaired. Despite the importance of skeletal muscle in regulating glucose homeostasis, the specific transcriptional changes associated with insulin-sensitive vs. -resistant states in muscle remain to be fully elucidated. Herein, using an RNA-seq approach we identified 20 genes differentially expressed in an insulin-resistant state in skeletal muscle, including cysteine- and glycine-rich protein 3 ( Csrp3), which was highly expressed in insulin-sensitive conditions but significantly reduced in the insulin-resistant state. CSRP3 has diverse functional roles including transcriptional regulation, signal transduction, and cytoskeletal organization, but its role in glucose homeostasis has yet to be explored. Thus, we investigated the role of CSRP3 in the development of obesity-induced insulin resistance in vivo. High-fat diet-fed CSRP3 knockout (KO) mice developed impaired glucose tolerance and insulin resistance as well as increased inflammation in skeletal muscle compared with wild-type (WT) mice. CSRP3-KO mice had significantly impaired insulin signaling, decreased GLUT4 translocation to the plasma membrane, and enhanced levels of phospho-PKCα in muscle, which all contributed to reduced insulin-stimulated glucose disposal in muscle in HFD-fed KO mice compared with WT mice. CSRP3 is a highly inducible protein and its expression is acutely increased after fasting. After 24h fasting, glucose tolerance was significantly improved in WT mice, but this effect was blunted in CSRP3-KO mice. In summary, we identify a novel role for Csrp3 expression in skeletal muscle in the development of obesity-induced insulin resistance.
Collapse
Affiliation(s)
| | - Natalie Weber
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Samuel A LaBarge
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, California
| | - Veronika Peterka
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Nhu Y Thi Doan
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Simon Schenk
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, California
| | - Olivia Osborn
- Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
27
|
Hypoxia promotes breast cancer cell invasion through HIF-1α-mediated up-regulation of the invadopodial actin bundling protein CSRP2. Sci Rep 2018; 8:10191. [PMID: 29976963 PMCID: PMC6033879 DOI: 10.1038/s41598-018-28637-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/13/2018] [Indexed: 12/20/2022] Open
Abstract
Hypoxia is a common feature of solid tumours that promotes invasion and metastatic dissemination. Invadopodia are actin-rich membrane protrusions that direct extracellular matrix proteolysis and facilitate tumour cell invasion. Here, we show that CSRP2, an invadopodial actin bundling protein, is upregulated by hypoxia in various breast cancer cell lines, as well as in pre-clinical and clinical breast tumour specimens. We functionally characterized two hypoxia responsive elements within the proximal promoter of CSRP2 gene which are targeted by hypoxia-inducible factor-1 (HIF-1) and required for promoter transactivation in response to hypoxia. Remarkably, CSRP2 knockdown significantly inhibits hypoxia-stimulated invadopodium formation, ECM degradation and invasion in MDA-MB-231 cells, while CSRP2 forced expression was sufficient to enhance the invasive capacity of HIF-1α-depleted cells under hypoxia. In MCF-7 cells, CSRP2 upregulation was required for hypoxia-induced formation of invadopodium precursors that were unable to promote ECM degradation. Collectively, our data support that CSRP2 is a novel and direct cytoskeletal target of HIF-1 which facilitates hypoxia-induced breast cancer cell invasion by promoting invadopodia formation.
Collapse
|
28
|
Wang SJ, Wang PZ, Gale RP, Qin YZ, Liu YR, Lai YY, Jiang H, Jiang Q, Zhang XH, Jiang B, Xu LP, Huang XJ, Liu KY, Ruan GR. Cysteine and glycine-rich protein 2 (CSRP2) transcript levels correlate with leukemia relapse and leukemia-free survival in adults with B-cell acute lymphoblastic leukemia and normal cytogenetics. Oncotarget 2018; 8:35984-36000. [PMID: 28415593 PMCID: PMC5482632 DOI: 10.18632/oncotarget.16416] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 03/11/2017] [Indexed: 12/14/2022] Open
Abstract
Relapse is the major cause of treatment-failure in adults with B-cell acute lymphoblastic leukemia (ALL) achieving complete remission after induction chemotherapy. Greater precision identifying persons likely to relapse is important. We did bio-informatics analyses of transcriptomic data to identify mRNA transcripts aberrantly-expressed in B-cell ALL. We selected 9 candidate genes for validation 7 of which proved significantly-associated with B-cell ALL. We next focused on function and clinical correlations of the cysteine and glycine-rich protein 2 (CSRP2). Quantitative real-time polymerase chain reaction (RT-qPCR) was used to examine gene transcript levels in bone marrow samples from 236 adults with B-cell ALL compared with samples from normals. CSRP2 was over-expressed in 228 out of 236 adults (97%) with newly-diagnosed B-cell ALL. A prognostic value was assessed in 168 subjects. In subjects with normal cytogenetics those with high CSRP2 transcript levels had a higher 5-year cumulative incidence of relapse (CIR) and worse relapse-free survival (RFS) compared with subjects with low transcript levels (56% [95% confidence interval, 53, 59%] vs. 19% [18, 20%]; P = 0.011 and 41% [17, 65%] vs. 80% [66–95%]; P = 0.007). In multivariate analyses a high CSRP2 transcript level was independently-associated with CIR (HR = 5.32 [1.64–17.28]; P = 0.005) and RFS (HR = 5.56 [1.87, 16.53]; P = 0.002). Functional analyses indicated CSRP2 promoted cell proliferation, cell-cycle progression, in vitro colony formation and cell migration ability. Abnormal CSRP2 expression was associated with resistance to chemotherapy; sensitivity was restored by down-regulating CSRP2 expression.
Collapse
Affiliation(s)
- Shu-Juan Wang
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Ping-Zhang Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Medical Immunology, Ministry of Health, China, Peking University Center for Human Disease Genomics, Beijing, China
| | - Robert Peter Gale
- Hematology Research Center, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, UK
| | - Ya-Zhen Qin
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yan-Rong Liu
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yue-Yun Lai
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Hao Jiang
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Qian Jiang
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Bin Jiang
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Kai-Yan Liu
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Guo-Rui Ruan
- Peking University People's Hospital and Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| |
Collapse
|
29
|
Campos LCG, Ribeiro-Silva JC, Menegon AS, Barauna VG, Miyakawa AA, Krieger JE. Cyclic stretch-induced Crp3 sensitizes vascular smooth muscle cells to apoptosis during vein arterialization remodeling. Clin Sci (Lond) 2018; 132:CS20171601. [PMID: 29437853 DOI: 10.1042/cs20171601] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/26/2018] [Accepted: 01/31/2018] [Indexed: 02/28/2024]
Abstract
Vein graft failure limits the long-term patency of the saphenous vein used as a conduit for coronary artery bypass graft. Early graft adaptation involves some degree of intima hyperplasia to sustain the hemodynamic stress, but the progress to occlusion in some veins remains unclear. We have demonstrated that stretch-induced up-regulation of cysteine and glycine-rich protein 3 (Crp3) in rat jugular vein and human saphenous vein in response to arterialization. Here, we developed a Crp3-KO rat to investigate the role of Crp3 in vascular remodeling. After 28 days jugular vein arterialization, the intima layer was 3-fold thicker in the Crp3-KO that showed comparable smooth muscle cells (SMC) proliferation but an absence of early apoptosis observed in the wild-type rat (WT). We then investigated the role of Crp3 in early integrin-mediated signaling apoptosis in isolated jugular SMC. Interestingly, under basal conditions, ceramide treatment failed to induce apoptosis in both WT and Crp3-KO SMC. Under stretch, Crp3 expression increased in WT SMC and ceramide induced apoptosis. Immunoblotting analysis indicated that ceramide stretch-induced apoptosis in SMC is accompanied by a decrease in the phosphorylation status of both Fak and Akt, leading to an increase in Bax expression and caspase-3 cleavage. In contrast, ceramide failed to decrease Fak and Akt phosphorylation in Crp3-KO SMC and, therefore, there was no downstream induction of Bax expression and effector caspase-3 cleavage. Taken together, we provide evidence that stretch-induced Crp3 modulates vein remodeling in response to arterialization by sensitizing SMC to apoptosis.
Collapse
Affiliation(s)
| | | | | | | | - Ayumi Aurea Miyakawa
- Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Jose Eduardo Krieger
- Department of Cardiopneumology, Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, 05403-000, Brazil
| |
Collapse
|
30
|
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: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
31
|
Hoffmann C, Mao X, Dieterle M, Moreau F, Al Absi A, Steinmetz A, Oudin A, Berchem G, Janji B, Thomas C. CRP2, a new invadopodia actin bundling factor critically promotes breast cancer cell invasion and metastasis. Oncotarget 2017; 7:13688-705. [PMID: 26883198 PMCID: PMC4924671 DOI: 10.18632/oncotarget.7327] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/27/2016] [Indexed: 01/11/2023] Open
Abstract
A critical process underlying cancer metastasis is the acquisition by tumor cells of an invasive phenotype. At the subcellular level, invasion is facilitated by actin-rich protrusions termed invadopodia, which direct extracellular matrix (ECM) degradation. Here, we report the identification of a new cytoskeletal component of breast cancer cell invadopodia, namely cysteine-rich protein 2 (CRP2). We found that CRP2 was not or only weakly expressed in epithelial breast cancer cells whereas it was up-regulated in mesenchymal/invasive breast cancer cells. In addition, high expression of the CRP2 encoding gene CSRP2 was associated with significantly increased risk of metastasis in basal-like breast cancer patients. CRP2 knockdown significantly reduced the invasive potential of aggressive breast cancer cells, whereas it did not impair 2D cell migration. In keeping with this, CRP2-depleted breast cancer cells exhibited a reduced capacity to promote ECM degradation, and to secrete and express MMP-9, a matrix metalloproteinase repeatedly associated with cancer progression and metastasis. In turn, ectopic expression of CRP2 in weakly invasive cells was sufficient to stimulate cell invasion. Both GFP-fused and endogenous CRP2 localized to the extended actin core of invadopodia, a structure primarily made of actin bundles. Purified recombinant CRP2 autonomously crosslinked actin filaments into thick bundles, suggesting that CRP2 contributes to the formation/maintenance of the actin core. Finally, CRP2 depletion significantly reduced the incidence of lung metastatic lesions in two xenograft mouse models of breast cancer. Collectively, our data identify CRP2 as a new cytoskeletal component of invadopodia that critically promotes breast cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Céline Hoffmann
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Xianqing Mao
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Monika Dieterle
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg.,NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Flora Moreau
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Antoun Al Absi
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - André Steinmetz
- Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Anaïs Oudin
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Guy Berchem
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Bassam Janji
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Clément Thomas
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
Collapse
|
32
|
Hoffmann C, Brown-Clay J, Thomas C. Subcellular localization and function of 2LIM proteins in plants and humans. PLANTA 2017; 246:1243-1245. [PMID: 28993895 DOI: 10.1007/s00425-017-2789-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 09/29/2017] [Indexed: 06/07/2023]
Affiliation(s)
- Céline Hoffmann
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, 84 Val, Fleuri, Luxembourg, Luxembourg
| | - Josh Brown-Clay
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, 84 Val, Fleuri, Luxembourg, Luxembourg
| | - Clément Thomas
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, 84 Val, Fleuri, Luxembourg, Luxembourg.
| |
Collapse
|
33
|
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: 1.0] [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]
|
34
|
Kihara T, Sugimoto Y, Shinohara S, Takaoka S, Miyake J. Cysteine-rich protein 2 accelerates actin filament cluster formation. PLoS One 2017; 12:e0183085. [PMID: 28813482 PMCID: PMC5558965 DOI: 10.1371/journal.pone.0183085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/29/2017] [Indexed: 12/22/2022] Open
Abstract
Filamentous actin (F-actin) forms many types of structures and dynamically regulates cell morphology and movement, and plays a mechanosensory role for extracellular stimuli. In this study, we determined that the smooth muscle-related transcription factor, cysteine-rich protein 2 (CRP2), regulates the supramolecular networks of F-actin. The structures of CRP2 and F-actin in solution were analyzed by small-angle X-ray solution scattering (SAXS). The general shape of CRP2 was partially unfolded and relatively ellipsoidal in structure, and the apparent cross sectional radius of gyration (Rc) was about 15.8 Å. The predicted shape, derived by ab initio modeling, consisted of roughly four tandem clusters: LIM domains were likely at both ends with the middle clusters being an unfolded linker region. From the SAXS analysis, the Rc of F-actin was about 26.7 Å, and it was independent of CRP2 addition. On the other hand, in the low angle region of the CRP2-bound F-actin scattering, the intensities showed upward curvature with the addition of CRP2, which indicates increasing branching of F-actin following CRP2 binding. From biochemical analysis, the actin filaments were augmented and clustered by the addition of CRP2. This F-actin clustering activity of CRP2 was cooperative with α-actinin. Thus, binding of CRP2 to F-actin accelerates actin polymerization and F-actin cluster formation.
Collapse
Affiliation(s)
- Takanori Kihara
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, Hibikino, Wakamatsu, Kitakyushu, Fukuoka, Japan
| | - Yasunobu Sugimoto
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi, Japan
| | - Satoko Shinohara
- Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Machikaneyama, Toyonaka, Osaka, Japan
| | - Shunpei Takaoka
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, Hibikino, Wakamatsu, Kitakyushu, Fukuoka, Japan
| | - Jun Miyake
- Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Machikaneyama, Toyonaka, Osaka, Japan
| |
Collapse
|
35
|
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: 0.9] [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
|
36
|
Lange S, Gehmlich K, Lun AS, Blondelle J, Hooper C, Dalton ND, Alvarez EA, Zhang X, Bang ML, Abassi YA, Dos Remedios CG, Peterson KL, Chen J, Ehler E. MLP and CARP are linked to chronic PKCα signalling in dilated cardiomyopathy. Nat Commun 2016; 7:12120. [PMID: 27353086 PMCID: PMC4931343 DOI: 10.1038/ncomms12120] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 05/31/2016] [Indexed: 11/28/2022] Open
Abstract
MLP (muscle LIM protein)-deficient mice count among the first mouse models for dilated cardiomyopathy (DCM), yet the exact role of MLP in cardiac signalling processes is still enigmatic. Elevated PKCα signalling activity is known to be an important contributor to heart failure. Here we show that MLP directly inhibits the activity of PKCα. In end-stage DCM, PKCα is concentrated at the intercalated disc of cardiomyocytes, where it is sequestered by the adaptor protein CARP in a multiprotein complex together with PLCβ1. In mice deficient for both MLP and CARP the chronic PKCα signalling chain at the intercalated disc is broken and they remain healthy. Our results suggest that the main role of MLP in heart lies in the direct inhibition of PKCα and that chronic uninhibited PKCα activity at the intercalated disc in the absence of functional MLP leads to heart failure. Altered function of the muscle LIM protein (MLP) causes dilated cardiomyopathy in mice and humans. Lange et al. explain the molecular role of MLP in the heart by showing that it affects the signalling complex at the intercalated discs of failing hearts that consists of PKCα, PLCβ1 and CARP by inhibiting PKCα auto-phosphorylation and function.
Collapse
Affiliation(s)
- Stephan Lange
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Katja Gehmlich
- BHF Centre of Research Excellence Oxford, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Alexander S Lun
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Jordan Blondelle
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Charlotte Hooper
- BHF Centre of Research Excellence Oxford, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Nancy D Dalton
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Erika A Alvarez
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Xiaoyu Zhang
- ACEA Biosciences, 6779 Mesa Ridge Rd #100, San Diego, CA-92121, USA
| | - Marie-Louise Bang
- Institute of Genetic and Biomedical Research, UOS Milan, National Research Council, Rozzano (Milan) 20089, Italy.,Humanitas Clinical and Research Center, Rozzano (Milan) 20089, Italy
| | - Yama A Abassi
- ACEA Biosciences, 6779 Mesa Ridge Rd #100, San Diego, CA-92121, USA
| | | | - Kirk L Peterson
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Ju Chen
- School of Medicine, University of California, San Diego, La Jolla CA-92093, USA
| | - Elisabeth Ehler
- BHF Centre of Research Excellence at King's College London, Cardiovascular Division and Randall Division of Cell and Molecular Biophysics, London SE1 1UL, UK
| |
Collapse
|
37
|
Bang ML. Animal Models of Congenital Cardiomyopathies Associated With Mutations in Z-Line Proteins. J Cell Physiol 2016; 232:38-52. [PMID: 27171814 DOI: 10.1002/jcp.25424] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/10/2016] [Indexed: 01/15/2023]
Abstract
The cardiac Z-line at the boundary between sarcomeres is a multiprotein complex connecting the contractile apparatus with the cytoskeleton and the extracellular matrix. The Z-line is important for efficient force generation and transmission as well as the maintenance of structural stability and integrity. Furthermore, it is a nodal point for intracellular signaling, in particular mechanosensing and mechanotransduction. Mutations in various genes encoding Z-line proteins have been associated with different cardiomyopathies, including dilated cardiomyopathy, hypertrophic cardiomyopathy, arrhythmogenic right ventricular cardiomyopathy, restrictive cardiomyopathy, and left ventricular noncompaction, and mutations even within the same gene can cause widely different pathologies. Animal models have contributed to a great advancement in the understanding of the physiological function of Z-line proteins and the pathways leading from mutations in Z-line proteins to cardiomyopathy, although genotype-phenotype prediction remains a great challenge. This review presents an overview of the currently available animal models for Z-line and Z-line associated proteins involved in human cardiomyopathies with special emphasis on knock-in and transgenic mouse models recapitulating the clinical phenotypes of human cardiomyopathy patients carrying mutations in Z-line proteins. Pros and cons of mouse models will be discussed and a future outlook will be given. J. Cell. Physiol. 232: 38-52, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research, UOS Milan, National Research Council and Humanitas Clinical and Research Center, Rozzano, Milan, Italy.
| |
Collapse
|
38
|
Zhu J, Bailly A, Zwiewka M, Sovero V, Di Donato M, Ge P, Oehri J, Aryal B, Hao P, Linnert M, Burgardt NI, Lücke C, Weiwad M, Michel M, Weiergräber OH, Pollmann S, Azzarello E, Mancuso S, Ferro N, Fukao Y, Hoffmann C, Wedlich-Söldner R, Friml J, Thomas C, Geisler M. TWISTED DWARF1 Mediates the Action of Auxin Transport Inhibitors on Actin Cytoskeleton Dynamics. THE PLANT CELL 2016; 28:930-48. [PMID: 27053424 PMCID: PMC4863381 DOI: 10.1105/tpc.15.00726] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 03/21/2016] [Accepted: 04/05/2016] [Indexed: 05/18/2023]
Abstract
Plant growth and architecture is regulated by the polar distribution of the hormone auxin. Polarity and flexibility of this process is provided by constant cycling of auxin transporter vesicles along actin filaments, coordinated by a positive auxin-actin feedback loop. Both polar auxin transport and vesicle cycling are inhibited by synthetic auxin transport inhibitors, such as 1-N-naphthylphthalamic acid (NPA), counteracting the effect of auxin; however, underlying targets and mechanisms are unclear. Using NMR, we map the NPA binding surface on the Arabidopsis thaliana ABCB chaperone TWISTED DWARF1 (TWD1). We identify ACTIN7 as a relevant, although likely indirect, TWD1 interactor, and show TWD1-dependent regulation of actin filament organization and dynamics and that TWD1 is required for NPA-mediated actin cytoskeleton remodeling. The TWD1-ACTIN7 axis controls plasma membrane presence of efflux transporters, and as a consequence act7 and twd1 share developmental and physiological phenotypes indicative of defects in auxin transport. These can be phenocopied by NPA treatment or by chemical actin (de)stabilization. We provide evidence that TWD1 determines downstream locations of auxin efflux transporters by adjusting actin filament debundling and dynamizing processes and mediating NPA action on the latter. This function appears to be evolutionary conserved since TWD1 expression in budding yeast alters actin polarization and cell polarity and provides NPA sensitivity.
Collapse
Affiliation(s)
- Jinsheng Zhu
- Department of Biology, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Aurelien Bailly
- Department of Biology, University of Fribourg, CH-1700 Fribourg, Switzerland Department of Plant and Microbial Biology, University of Zurich, CH-8008 Zurich, Switzerland
| | - Marta Zwiewka
- CEITEC-Central European Institute of Technology, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Valpuri Sovero
- Department of Plant and Microbial Biology, University of Zurich, CH-8008 Zurich, Switzerland
| | - Martin Di Donato
- Department of Biology, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Pei Ge
- Department of Biology, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Jacqueline Oehri
- Department of Biology, University of Fribourg, CH-1700 Fribourg, Switzerland Institute of Evolutionary Biology and Environmental Studies, University of Zurich, CH-8057 Zurich, Switzerland
| | - Bibek Aryal
- Department of Biology, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Pengchao Hao
- Department of Biology, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Miriam Linnert
- Max Planck Research Unit for Enzymology of Protein Folding, D-06099 Halle (Saale), Germany
| | - Noelia Inés Burgardt
- Max Planck Research Unit for Enzymology of Protein Folding, D-06099 Halle (Saale), Germany Institute of Biochemistry and Biophysics (IQUIFIB), School of Pharmacy and Biochemistry, University of Buenos Aires, C1113AAD Buenos Aires, Argentina
| | - Christian Lücke
- Max Planck Research Unit for Enzymology of Protein Folding, D-06099 Halle (Saale), Germany
| | - Matthias Weiwad
- Max Planck Research Unit for Enzymology of Protein Folding, D-06099 Halle (Saale), Germany Department of Enzymology, Martin-Luther-University Halle-Wittenberg, Institute of Biochemistry and Biotechnology, D-06099 Halle, Germany
| | - Max Michel
- Institute of Complex Systems, ICS-6: Structural Biochemistry, D-52425 Jülich, Germany
| | - Oliver H Weiergräber
- Institute of Complex Systems, ICS-6: Structural Biochemistry, D-52425 Jülich, Germany
| | - Stephan Pollmann
- Centro de Biotecnología y Genómica de Plantas, 28223 Pozuelo de Alarcón, Madrid, Spain
| | | | | | - Noel Ferro
- University of Bonn, Mulliken Center for Theoretical Chemistry, Institute for Physical and Theoretical Chemistry, D-53115 Bonn, Germany
| | - Yoichiro Fukao
- Plant Global Educational Project, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| | - Céline Hoffmann
- Cytoskeleton and Cancer Progression, Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | | | - Jiří Friml
- Institute of Science and Technology Austria, A-3400 Klosterneuburg, Austria
| | - Clément Thomas
- Cytoskeleton and Cancer Progression, Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Markus Geisler
- Department of Biology, University of Fribourg, CH-1700 Fribourg, Switzerland Department of Plant and Microbial Biology, University of Zurich, CH-8008 Zurich, Switzerland
| |
Collapse
|
39
|
New insights into the protein aggregation pathology in myotilinopathy by combined proteomic and immunolocalization analyses. Acta Neuropathol Commun 2016; 4:8. [PMID: 26842778 PMCID: PMC4739336 DOI: 10.1186/s40478-016-0280-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 01/23/2016] [Indexed: 01/09/2023] Open
Abstract
Introduction Myofibrillar myopathies are characterized by progressive muscle weakness and impressive abnormal protein aggregation in muscle fibers. In about 10 % of patients, the disease is caused by mutations in the MYOT gene encoding myotilin. The aim of our study was to decipher the composition of protein deposits in myotilinopathy to get new information about aggregate pathology. Results Skeletal muscle samples from 15 myotilinopathy patients were included in the study. Aggregate and control samples were collected from muscle sections by laser microdissection and subsequently analyzed by a highly sensitive proteomic approach that enables a relative protein quantification. In total 1002 different proteins were detected. Seventy-six proteins showed a significant over-representation in aggregate samples including 66 newly identified aggregate proteins. Z-disc-associated proteins were the most abundant aggregate components, followed by sarcolemmal and extracellular matrix proteins, proteins involved in protein quality control and degradation, and proteins with a function in actin dynamics or cytoskeletal transport. Forty over-represented proteins were evaluated by immunolocalization studies. These analyses validated our mass spectrometric data and revealed different regions of protein accumulation in abnormal muscle fibers. Comparison of data from our proteomic analysis in myotilinopathy with findings in other myofibrillar myopathy subtypes indicates a characteristic basic pattern of aggregate composition and resulted in identification of a highly sensitive and specific diagnostic marker for myotilinopathy. Conclusions Our findings i) indicate that main protein components of aggregates belong to a network of interacting proteins, ii) provide new insights into the complex regulation of protein degradation in myotilinopathy that may be relevant for new treatment strategies, iii) imply a combination of a toxic gain-of-function leading to myotilin-positive protein aggregates and a loss-of-function caused by a shift in subcellular distribution with a deficiency of myotilin at Z-discs that impairs the integrity of myofibrils, and iv) demonstrate that proteomic analysis can be helpful in differential diagnosis of protein aggregate myopathies. Electronic supplementary material The online version of this article (doi:10.1186/s40478-016-0280-0) contains supplementary material, which is available to authorized users.
Collapse
|
40
|
Moes D, Hoffmann C, Dieterle M, Moreau F, Neumann K, Papuga J, Furtado AT, Steinmetz A, Thomas C. The pH sensibility of actin-bundling LIM proteins is governed by the acidic properties of their C-terminal domain. FEBS Lett 2015; 589:2312-9. [PMID: 26226417 DOI: 10.1016/j.febslet.2015.07.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/17/2015] [Accepted: 07/17/2015] [Indexed: 11/22/2022]
Abstract
Actin-bundling Arabidopsis LIM proteins are subdivided into two subfamilies differing in their pH sensitivity. Widely-expressed WLIMs are active under low and high physiologically-relevant pH conditions, whereas pollen-enriched PLIMs are inactivated by pH values above 6.8. By a domain swapping approach we identified the C-terminal (Ct) domain of PLIMs as the domain responsible for pH responsiveness. Remarkably, this domain conferred pH sensitivity to LIM proteins, when provided "in trans" (i.e., as a single, independent, peptide), indicating that it operates through the interaction with another domain. An acidic 6xc-Myc peptide functionally mimicked the Ct domain of PLIMs and efficiently inhibited LIM actin bundling activity under high pH conditions. Together, our data suggest a model where PLIMs are regulated by an intermolecular interaction between their acidic Ct domain and another, yet unidentified, domain.
Collapse
Affiliation(s)
- Danièle Moes
- Department of Oncology, Luxembourg Institute of Health (L.I.H.), L-1526 Luxembourg, Luxembourg
| | - Céline Hoffmann
- Department of Oncology, Luxembourg Institute of Health (L.I.H.), L-1526 Luxembourg, Luxembourg
| | - Monika Dieterle
- Department of Oncology, Luxembourg Institute of Health (L.I.H.), L-1526 Luxembourg, Luxembourg
| | - Flora Moreau
- Department of Oncology, Luxembourg Institute of Health (L.I.H.), L-1526 Luxembourg, Luxembourg
| | - Katrin Neumann
- Department of Oncology, Luxembourg Institute of Health (L.I.H.), L-1526 Luxembourg, Luxembourg
| | - Jessica Papuga
- Department of Oncology, Luxembourg Institute of Health (L.I.H.), L-1526 Luxembourg, Luxembourg
| | - Angela Tavares Furtado
- Department of Oncology, Luxembourg Institute of Health (L.I.H.), L-1526 Luxembourg, Luxembourg
| | - André Steinmetz
- Department of Oncology, Luxembourg Institute of Health (L.I.H.), L-1526 Luxembourg, Luxembourg
| | - Clément Thomas
- Department of Oncology, Luxembourg Institute of Health (L.I.H.), L-1526 Luxembourg, Luxembourg.
| |
Collapse
|
41
|
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: 58] [Impact Index Per Article: 6.4] [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
|
42
|
XIRP2, an actin-binding protein essential for inner ear hair-cell stereocilia. Cell Rep 2015; 10:1811-8. [PMID: 25772365 DOI: 10.1016/j.celrep.2015.02.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/30/2015] [Accepted: 02/14/2015] [Indexed: 01/20/2023] Open
Abstract
Hair cells of the inner ear are mechanoreceptors for hearing and balance, and proteins highly enriched in hair cells may have specific roles in the development and maintenance of the mechanotransduction apparatus. We identified XIRP2/mXinβ as an enriched protein likely to be essential for hair cells. We found that different isoforms of this protein are expressed and differentially located: short splice forms (also called XEPLIN) are targeted more to stereocilia, whereas two long isoforms containing a XIN-repeat domain are in both stereocilia and cuticular plates. Mice lacking the Xirp2 gene developed normal stereocilia bundles, but these degenerated with time: stereocilia were lost and long membranous protrusions emanated from the nearby apical surfaces. At an ultrastructural level, the paracrystalline actin filaments became disorganized. XIRP2 is apparently involved in the maintenance of actin structures in stereocilia and cuticular plates of hair cells, and perhaps in other organs where it is expressed.
Collapse
|