1
|
Xue S, Benvie AM, Blum JE, Kolba NJ, Cosgrove BD, Thalacker-Mercer A, Berry DC. Suppressing PDGFRβ Signaling Enhances Myocyte Fusion to Promote Skeletal Muscle Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618247. [PMID: 39464006 PMCID: PMC11507758 DOI: 10.1101/2024.10.15.618247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Muscle cell fusion is critical for forming and maintaining multinucleated myotubes during skeletal muscle development and regeneration. However, the molecular mechanisms directing cell-cell fusion are not fully understood. Here, we identify platelet-derived growth factor receptor beta (PDGFRβ) signaling as a key modulator of myocyte fusion in adult muscle cells. Our findings demonstrate that genetic deletion of Pdgfrβ enhances muscle regeneration and increases myofiber size, whereas PDGFRβ activation impairs muscle repair. Inhibition of PDGFRβ activity promotes myonuclear accretion in both mouse and human myotubes, whereas PDGFRβ activation stalls myotube development by preventing cell spreading to limit fusion potential. Transcriptomics analysis show that PDGFRβ signaling cooperates with TGFβ signaling to direct myocyte size and fusion. Mechanistically, PDGFRβ signaling requires STAT1 activation, and blocking STAT1 phosphorylation enhances myofiber repair and size during regeneration. Collectively, PDGFRβ signaling acts as a regenerative checkpoint and represents a potential clinical target to rapidly boost skeletal muscle repair.
Collapse
Affiliation(s)
- Siwen Xue
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | - Abigail M Benvie
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | - Jamie E Blum
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Current address: Department of Chemical Engineering; Stanford University; Stanford, CA
| | - Nikolai J Kolba
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | | | - Anna Thalacker-Mercer
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Department of Cell, Development and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel C Berry
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Corresponding author
| |
Collapse
|
2
|
Archacka K, Grabowska I, Mierzejewski B, Graffstein J, Górzyńska A, Krawczyk M, Różycka AM, Kalaszczyńska I, Muras G, Stremińska W, Jańczyk-Ilach K, Walczak P, Janowski M, Ciemerych MA, Brzoska E. Hypoxia preconditioned bone marrow-derived mesenchymal stromal/stem cells enhance myoblast fusion and skeletal muscle regeneration. Stem Cell Res Ther 2021; 12:448. [PMID: 34372911 PMCID: PMC8351116 DOI: 10.1186/s13287-021-02530-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022] Open
Abstract
Background The skeletal muscle reconstruction occurs thanks to unipotent stem cells, i.e., satellite cells. The satellite cells remain quiescent and localized between myofiber sarcolemma and basal lamina. They are activated in response to muscle injury, proliferate, differentiate into myoblasts, and recreate myofibers. The stem and progenitor cells support skeletal muscle regeneration, which could be disturbed by extensive damage, sarcopenia, cachexia, or genetic diseases like dystrophy. Many lines of evidence showed that the level of oxygen regulates the course of cell proliferation and differentiation. Methods In the present study, we analyzed hypoxia impact on human and pig bone marrow-derived mesenchymal stromal cell (MSC) and mouse myoblast proliferation, differentiation, and fusion. Moreover, the influence of the transplantation of human bone marrow-derived MSCs cultured under hypoxic conditions on skeletal muscle regeneration was studied. Results We showed that bone marrow-derived MSCs increased VEGF expression and improved myogenesis under hypoxic conditions in vitro. Transplantation of hypoxia preconditioned bone marrow-derived MSCs into injured muscles resulted in the improved cell engraftment and formation of new vessels. Conclusions We suggested that SDF-1 and VEGF secreted by hypoxia preconditioned bone marrow-derived MSCs played an essential role in cell engraftment and angiogenesis. Importantly, hypoxia preconditioned bone marrow-derived MSCs more efficiently engrafted injured muscles; however, they did not undergo myogenic differentiation. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02530-3.
Collapse
Affiliation(s)
- Karolina Archacka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Bartosz Mierzejewski
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Joanna Graffstein
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Alicja Górzyńska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Marta Krawczyk
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Anna M Różycka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Ilona Kalaszczyńska
- Department of Histology and Embryology, Medical University of Warsaw, 02-004, Warsaw, Poland.,Laboratory for Cell Research and Application, Medical University of Warsaw, 02-097, Warsaw, Poland
| | - Gabriela Muras
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Władysława Stremińska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Katarzyna Jańczyk-Ilach
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Piotr Walczak
- Department of Pathophysiology, Faculty of Medical Sciences, University of Warmia and Mazury, Warszawska 30 St, 10-082, Olsztyn, Poland.,Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mirosław Janowski
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, 21201, USA.,NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5 St, 02-106, Warsaw, Poland
| | - Maria A Ciemerych
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland.
| |
Collapse
|
3
|
Kim Y, Yang DS, Katti P, Glancy B. Protein composition of the muscle mitochondrial reticulum during postnatal development. J Physiol 2019; 597:2707-2727. [PMID: 30919448 PMCID: PMC6826232 DOI: 10.1113/jp277579] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/11/2019] [Indexed: 01/24/2023] Open
Abstract
KEY POINTS Muscle mitochondrial networks changed from a longitudinal, fibre parallel orientation to a perpendicular configuration during postnatal development. Mitochondrial dynamics, mitophagy and calcium uptake proteins were abundant during early postnatal development. Mitochondrial biogenesis and oxidative phosphorylation proteins were upregulated throughout muscle development. Postnatal muscle mitochondrial network formation is accompanied by a change in protein expression profile from mitochondria designed for co-ordinated cellular assembly to mitochondria highly specialized for cellular energy metabolism. ABSTRACT Striated muscle mitochondria form connected networks capable of rapid cellular energy distribution. However, the mitochondrial reticulum is not formed at birth and the mechanisms driving network development remain unclear. In the present study, we aimed to establish the network formation timecourse and protein expression profile during postnatal development of the murine muscle mitochondrial reticulum. Two-photon microscopy was used to observe mitochondrial network orientation in tibialis anterior (TA) muscles of live mice at postnatal days (P) 1, 7, 14, 21 and 42, respectively. All muscle fibres maintained a longitudinal, fibre parallel mitochondrial network orientation early in development (P1-7). Mixed networks were most common at P14 but, by P21, almost all fibres had developed the perpendicular mitochondrial orientation observed in mature, glycolytic fibres. Tandem mass tag proteomics were then applied to examine changes in 6869 protein abundances in developing TA muscles. Mitochondrial proteins increased by 32% from P1 to P42. In addition, both nuclear- and mitochondrial-DNA encoded oxidative phosphorylation (OxPhos) components were increased during development, whereas OxPhos assembly factors decreased. Although mitochondrial dynamics and mitophagy were induced at P1-7, mitochondrial biogenesis was enhanced after P14. Moreover, calcium signalling proteins and the mitochondrial calcium uniporter had the highest expression early in postnatal development. In conclusion, mitochondrial networks transform from a fibre parallel to perpendicular orientation during the second and third weeks after birth in murine glycolytic skeletal muscle. This structural transition is accompanied by a change in protein expression profile from mitochondria designed for co-ordinated cellular assembly to mitochondria highly specialized for cellular energy metabolism.
Collapse
Affiliation(s)
- Yuho Kim
- National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaMDUSA
| | - Daniel S. Yang
- National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaMDUSA
| | - Prasanna Katti
- National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaMDUSA
| | - Brian Glancy
- National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaMDUSA
- National Institute of Arthritis and Musculoskeletal and Skin DiseasesNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
4
|
Becker A, Zhang P, Allmann L, Meilinger D, Bertulat B, Eck D, Hofstaetter M, Bartolomei G, Hottiger MO, Schreiber V, Leonhardt H, Cardoso MC. Poly(ADP-ribosyl)ation of Methyl CpG Binding Domain Protein 2 Regulates Chromatin Structure. J Biol Chem 2016; 291:4873-81. [PMID: 26772194 PMCID: PMC4777825 DOI: 10.1074/jbc.m115.698357] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Indexed: 11/06/2022] Open
Abstract
The epigenetic information encoded in the genomic DNA methylation pattern is translated by methylcytosine binding proteins like MeCP2 into chromatin topology and structure and gene activity states. We have shown previously that the MeCP2 level increases during differentiation and that it causes large-scale chromatin reorganization, which is disturbed by MeCP2 Rett syndrome mutations. Phosphorylation and other posttranslational modifications of MeCP2 have been described recently to modulate its function. Here we show poly(ADP-ribosyl)ation of endogenous MeCP2 in mouse brain tissue. Consequently, we found that MeCP2 induced aggregation of pericentric heterochromatin and that its chromatin accumulation was enhanced in poly(ADP-ribose) polymerase (PARP) 1(-/-) compared with wild-type cells. We mapped the poly(ADP-ribosyl)ation domains and engineered MeCP2 mutation constructs to further analyze potential effects on DNA binding affinity and large-scale chromatin remodeling. Single or double deletion of the poly(ADP-ribosyl)ated regions and PARP inhibition increased the heterochromatin clustering ability of MeCP2. Increased chromatin clustering may reflect increased binding affinity. In agreement with this hypothesis, we found that PARP-1 deficiency significantly increased the chromatin binding affinity of MeCP2 in vivo. These data provide novel mechanistic insights into the regulation of MeCP2-mediated, higher-order chromatin architecture and suggest therapeutic opportunities to manipulate MeCP2 function.
Collapse
Affiliation(s)
- Annette Becker
- From the Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Peng Zhang
- From the Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Lena Allmann
- From the Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Daniela Meilinger
- the Center for Integrated Protein Science at the Department of Biology, Ludwig Maximilians University Munich, 82152 Planegg-Martinsried, Germany
| | - Bianca Bertulat
- From the Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Daniel Eck
- From the Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Maria Hofstaetter
- the Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Giody Bartolomei
- the Department of Molecular Mechanisms of Disease, University of Zurich, 8057 Zurich, Switzerland, and
| | - Michael O Hottiger
- the Department of Molecular Mechanisms of Disease, University of Zurich, 8057 Zurich, Switzerland, and
| | - Valérie Schreiber
- UMR7242 Biotechnology and Cell Signaling, Laboratory of Excellence Medalis, Strasbourg University, CNRS, Ecole Superieure de Biotechnologie de Strasbourg, BP10413, 67412 Illkirch Cedex, France
| | - Heinrich Leonhardt
- the Center for Integrated Protein Science at the Department of Biology, Ludwig Maximilians University Munich, 82152 Planegg-Martinsried, Germany
| | - M Cristina Cardoso
- From the Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany,
| |
Collapse
|
5
|
Ectopic expression of Msx2 in mammalian myotubes recapitulates aspects of amphibian muscle dedifferentiation. Stem Cell Res 2015; 15:542-553. [DOI: 10.1016/j.scr.2015.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 09/23/2015] [Accepted: 09/26/2015] [Indexed: 11/20/2022] Open
|
6
|
Jost KL, Bertulat B, Rapp A, Brero A, Hardt T, Domaing P, Gösele C, Schulz H, Hübner N, Cardoso MC. Gene repositioning within the cell nucleus is not random and is determined by its genomic neighborhood. Epigenetics Chromatin 2015; 8:36. [PMID: 26388944 PMCID: PMC4574441 DOI: 10.1186/s13072-015-0025-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/01/2015] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Heterochromatin has been reported to be a major silencing compartment during development and differentiation. Prominent heterochromatin compartments are located at the nuclear periphery and inside the nucleus (e.g., pericentric heterochromatin). Whether the position of a gene in relation to some or all heterochromatin compartments matters remains a matter of debate, which we have addressed in this study. Answering this question demanded solving the technical challenges of 3D measurements and the large-scale morphological changes accompanying cellular differentiation. RESULTS Here, we investigated the proximity effects of the nuclear periphery and pericentric heterochromatin on gene expression and additionally considered the effect of neighboring genomic features on a gene's nuclear position. Using a well-established myogenic in vitro differentiation system and a differentiation-independent heterochromatin remodeling system dependent on ectopic MeCP2 expression, we first identified genes with statistically significant expression changes by transcriptional profiling. We identified nuclear gene positions by 3D fluorescence in situ hybridization followed by 3D distance measurements toward constitutive and facultative heterochromatin domains. Single-cell-based normalization enabled us to acquire morphologically unbiased data and we finally correlated changes in gene positioning to changes in transcriptional profiles. We found no significant correlation of gene silencing and proximity to constitutive heterochromatin and a rather unexpected inverse correlation of gene activity and position relative to facultative heterochromatin at the nuclear periphery. CONCLUSION In summary, our data question the hypothesis of heterochromatin as a general silencing compartment. Nonetheless, compared to a simulated random distribution, we found that genes are not randomly located within the nucleus. An analysis of neighboring genomic context revealed that gene location within the nucleus is rather dependent on CpG islands, GC content, gene density, and short and long interspersed nuclear elements, collectively known as RIDGE (regions of increased gene expression) properties. Although genes do not move away/to the heterochromatin upon up-/down-regulation, genomic regions with RIDGE properties are generally excluded from peripheral heterochromatin. Hence, we suggest that individual gene activity does not influence gene positioning, but rather chromosomal context matters for sub-nuclear location.
Collapse
Affiliation(s)
- K Laurence Jost
- Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Bianca Bertulat
- Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Alexander Rapp
- Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Alessandro Brero
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Tanja Hardt
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Petra Domaing
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Claudia Gösele
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Herbert Schulz
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Norbert Hübner
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - M Cristina Cardoso
- Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| |
Collapse
|
7
|
WNT/β-Catenin Signaling Regulates Multiple Steps of Myogenesis by Regulating Step-Specific Targets. Mol Cell Biol 2015; 35:1763-76. [PMID: 25755281 DOI: 10.1128/mcb.01180-14] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 02/27/2015] [Indexed: 12/23/2022] Open
Abstract
Molecules involved in WNT/β-catenin signaling show specific spatiotemporal expression and play vital roles in myogenesis; however, it is still largely unknown how WNT/β-catenin signaling regulates each step of myogenesis. Here, we show that WNT/β-catenin signaling can control diverse biological processes of myogenesis by regulating step-specific molecules. In order to identify the temporally specific roles of WNT/β-catenin signaling molecules in muscle development and homeostasis, we used in vitro culture systems for both primary mouse myoblasts and C2C12 cells, which can differentiate into myofibers. We found that a blockade of WNT/β-catenin signaling in the proliferating cells decreases proliferation activity, but does not induce cell death, through the regulation of genes cyclin A2 (Ccna2) and cell division cycle 25C (Cdc25c). During muscle differentiation, the inhibition of WNT/β-catenin signaling blocks myoblast fusion through the inhibition of the Fermitin family homolog 2 (Fermt2) gene. Blocking WNT/β-catenin signaling in the well-differentiated myofibers results in the failure of maintenance of their structure by disruption of cadherin/β-catenin/actin complex formation, which plays a crucial role in connecting a myofiber's cytoskeleton to the surrounding extracellular matrix. Thus, our results indicate that WNT/β-catenin signaling can regulate multiple steps of myogenesis, including cell proliferation, myoblast fusion, and homeostasis, by targeting step-specific molecules.
Collapse
|
8
|
Marti M, Montserrat N, Pardo C, Mulero L, Miquel-Serra L, Rodrigues AMC, Andrés Vaquero J, Kuebler B, Morera C, Barrero MJ, Izpisua Belmonte JC. M-cadherin-mediated intercellular interactions activate satellite cell division. J Cell Sci 2013; 126:5116-31. [PMID: 24046443 DOI: 10.1242/jcs.123562] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Adult muscle stem cells and their committed myogenic precursors, commonly referred to as the satellite cell population, are involved in both muscle growth after birth and regeneration after damage. It has been previously proposed that, under these circumstances, satellite cells first become activated, divide and differentiate, and only later fuse to the existing myofiber through M-cadherin-mediated intercellular interactions. Our data show that satellite cells fuse with the myofiber concomitantly to cell division, and only when the nuclei of the daughter cells are inside the myofiber, do they complete the process of differentiation. Here we demonstrate that M-cadherin plays an important role in cell-to-cell recognition and fusion, and is crucial for cell division activation. Treatment of satellite cells with M-cadherin in vitro stimulates cell division, whereas addition of anti-M-cadherin antibodies reduces the cell division rate. Our results suggest an alternative model for the contribution of satellite cells to muscle development, which might be useful in understanding muscle regeneration, as well as muscle-related dystrophies.
Collapse
Affiliation(s)
- Merce Marti
- Center of Regenerative Medicine in Barcelona, Dr. Aiguader, 88, 08003 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Adhesion proteins--an impact on skeletal myoblast differentiation. PLoS One 2013; 8:e61760. [PMID: 23671573 PMCID: PMC3645998 DOI: 10.1371/journal.pone.0061760] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 03/13/2013] [Indexed: 11/19/2022] Open
Abstract
Formation of mammalian skeletal muscle myofibers, that takes place during embryogenesis, muscle growth or regeneration, requires precise regulation of myoblast adhesion and fusion. There are few evidences showing that adhesion proteins play important role in both processes. To follow the function of these molecules in myoblast differentiation we analysed integrin alpha3, integrin beta1, ADAM12, CD9, CD81, M-cadherin, and VCAM-1 during muscle regeneration. We showed that increase in the expression of these proteins accompanies myoblast fusion and myotube formation in vivo. We also showed that during myoblast fusion in vitro integrin alpha3 associates with integrin beta1 and ADAM12, and also CD9 and CD81, but not with M-cadherin or VCAM-1. Moreover, we documented that experimental modification in the expression of integrin alpha3 lead to the modification of myoblast fusion in vitro. Underexpression of integrin alpha3 decreased myoblasts' ability to fuse. This phenomenon was not related to the modifications in the expression of other adhesion proteins, i.e. integrin beta1, CD9, CD81, ADAM12, M-cadherin, or VCAM-1. Apparently, aberrant expression only of one partner of multiprotein adhesion complexes necessary for myoblast fusion, in this case integrin alpha3, prevents its proper function. Summarizing, we demonstrated the importance of analysed adhesion proteins in myoblast fusion both in vivo and in vitro.
Collapse
|
10
|
Lee EJ, Pokharel S, Kim JH, Nam SS, Choi IH. Effect of Gender-Specific Adult Bovine Serum on Gene Expression During Myogenesis. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2012. [DOI: 10.5187/jast.2012.54.3.219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
11
|
Ceccarelli G, Benedetti L, Galli D, Prè D, Silvani G, Crosetto N, Magenes G, Cusella De Angelis MG. Low-amplitude high frequency vibration down-regulates myostatin and atrogin-1 expression, two components of the atrophy pathway in muscle cells. J Tissue Eng Regen Med 2012; 8:396-406. [PMID: 22711460 DOI: 10.1002/term.1533] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 01/18/2012] [Accepted: 04/04/2012] [Indexed: 11/07/2022]
Abstract
Whole body vibration (WBV) is a very widespread mechanical stimulus used in physical therapy, rehabilitation and fitness centres. It has been demonstrated that vibration induces improvements in muscular strength and performance and increases bone density. We investigated the effects of low-amplitude, high frequency vibration (HFV) at the cellular and tissue levels in muscle. We developed a system to produce vibrations adapted to test several parameters in vitro and in vivo. For in vivo experiments, we used newborn CD1 wild-type mice, for in vitro experiments, we isolated satellite cells from 6-day-old CD1 mice, while for proliferation studies, we used murine cell lines. Animals and cells were treated with high frequency vibration at 30 Hz. We analyzed the effects of mechanical stimulation on muscle hypertrophy/atrophy pathways, fusion enhancement of myoblast cells and modifications in the proliferation rate of cells. Results demonstrated that mechanical vibration strongly down-regulates atrophy genes both in vivo and in vitro. The in vitro experiments indicated that mechanical stimulation promotes fusion of satellite cells treated directly in culture compared to controls. Finally, proliferation experiments indicated that stimulated cells had a decreased growth rate compared to controls. We concluded that vibration treatment at 30 Hz is effective in suppressing the atrophy pathway both in vivo and in vitro and enhances fusion of satellite muscle cells.
Collapse
Affiliation(s)
- Gabriele Ceccarelli
- Dipartimento di Medicina Sperimentale, University of Pavia, Italy; Centro di Ingegneria Tissutale, University of Pavia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Tanaka H, Takafuji K, Taguchi A, Wiriyasermkul P, Ohgaki R, Nagamori S, Suh PG, Kanai Y. Linkage of N-cadherin to multiple cytoskeletal elements revealed by a proteomic approach in hippocampal neurons. Neurochem Int 2012; 61:240-50. [PMID: 22609377 DOI: 10.1016/j.neuint.2012.05.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 05/07/2012] [Accepted: 05/09/2012] [Indexed: 01/21/2023]
Abstract
The CNS synapse is an adhesive junction differentiated for chemical neurotransmission and is equipped with presynaptic vesicles and postsynaptic neurotransmitter receptors. Cell adhesion molecule cadherins not only maintain connections between pre- and postsynaptic membranes but also modulate the efficacy of synaptic transmission. Although the components of the cadherin-mediated adhesive apparatus have been studied extensively in various cell systems, the complete picture of these components, particularly at the synaptic junction, remains elusive. Here, we describe the proteomic assortment of the N-cadherin-mediated synaptic adhesion apparatus in cultured hippocampal neurons. N-cadherin immunoprecipitated from Triton X-100-solubilized neuronal extract contained equal amounts of β- and α-catenins, as well as F-actin-related membrane anchor proteins such as integrins bridged with α-actinin-4, and Na(+)/K(+)-ATPase bridged with spectrins. A close relative of β-catenin, plakoglobin, and its binding partner, desmoplakin, were also found, suggesting that a subset of the N-cadherin-mediated adhesive apparatus also anchors intermediate filaments. Moreover, dynein heavy chain and LEK1/CENPF/mitosin were found. This suggests that internalized pools of N-cadherin in trafficking vesicles are conveyed by dynein motors on microtubules. In addition, ARVCF and NPRAP/neurojungin/δ2-catenin, but not p120ctn/δ1-catenin or plakophilins-1, -2, -3, -4 (p0071), were found, suggesting other possible bridges to microtubules. Finally, synaptic stimulation by membrane depolarization resulted in an increased 93-kDa band, which corresponded to proteolytically truncated β-catenin. The integration of three different classes of cytoskeletal systems found in the synaptic N-cadherin complex may imply a dynamic switching of adhesive scaffolds in response to synaptic activity.
Collapse
Affiliation(s)
- Hidekazu Tanaka
- Department of Pharmacology, Osaka University School of Medicine, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Agarwal N, Becker A, Jost KL, Haase S, Thakur BK, Brero A, Hardt T, Kudo S, Leonhardt H, Cardoso MC. MeCP2 Rett mutations affect large scale chromatin organization. Hum Mol Genet 2011; 20:4187-95. [DOI: 10.1093/hmg/ddr346] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
14
|
Green YS, Vetter ML. EBF proteins participate in transcriptional regulation of Xenopus muscle development. Dev Biol 2011; 358:240-50. [PMID: 21839736 DOI: 10.1016/j.ydbio.2011.07.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 07/24/2011] [Accepted: 07/27/2011] [Indexed: 01/08/2023]
Abstract
EBF proteins have diverse functions in the development of multiple lineages, including neurons, B cells and adipocytes. During Drosophila muscle development EBF proteins are expressed in muscle progenitors and are required for muscle cell differentiation, but there is no known function of EBF proteins in vertebrate muscle development. In this study, we examine the expression of ebf genes in Xenopus muscle tissue and show that EBF activity is necessary for aspects of Xenopus skeletal muscle development, including somite organization, migration of hypaxial muscle anlagen toward the ventral abdomen, and development of jaw muscle. From a microarray screen, we have identified multiple candidate targets of EBF activity with known roles in muscle development. The candidate targets we have verified are MYOD, MYF5, M-Cadherin and SEB-4. In vivo overexpression of the ebf2 and ebf3 genes leads to ectopic expression of these candidate targets, and knockdown of EBF activity causes downregulation of the endogenous expression of the candidate targets. Furthermore, we found that MYOD and MYF5 are likely to be direct targets. Finally we show that MYOD can upregulate the expression of ebf genes, indicating the presence of a positive feedback loop between EBF and MYOD that we find to be important for maintenance of MYOD expression in Xenopus. These results suggest that EBF activity is important for both stabilizing commitment and driving aspects of differentiation in Xenopus muscle cells.
Collapse
Affiliation(s)
- Yangsook Song Green
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | | |
Collapse
|
15
|
Comparison of satellite cell-derived myoblasts and C2C12 differentiation in two- and three-dimensional cultures: changes in adhesion protein expression. Cell Biol Int 2011; 35:125-33. [PMID: 20836763 DOI: 10.1042/cbi20090335] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Changes in the expression of adhesion proteins involved in myoblast differentiation were investigated in monolayer (two-dimensional) and 3D (three-dimensional) cell cultures. The expression of integrin alpha3 subunit, integrin beta1 subunit, ADAM12 (a disintegrin and metalloproteinase 12), tetraspanins CD9 and CD81 and M-cadherin were examined in the murine myoblast cell line C2C12 and in a primary culture of rat satellite cells. Myoblasts in monolayer and 3D cultures showed significant differences in their morphology and cytoskeletal organization. All of the studied proteins participated in myoblast fusion in each culture examined, but differences in their levels of expression were observed. Satellite cell-derived myoblasts exhibited higher expression of adhesion protein mRNAs than C2C12 cells. Also, C2C12 cells from a 3D culture showed slightly higher expression of adhesion protein transcripts than the same cells cultured as a monolayer. Significantly, the levels of adhesion protein mRNAs were found to change in parallel in all cell culture types. Despite this finding, it is important that differences between satellite cell-derived myoblasts and cell line C2C12 grown in monolayer and 3D cultures are taken into account when studying processes of myoblast differentiation in vitro.
Collapse
|
16
|
Balan OV, Ozernuk ND. Expression of the gene MyoD and m-cadherin in the myogenic precursor cell culture isolated from muscles of rats at different stages of ontogenesis. BIOL BULL+ 2011. [DOI: 10.1134/s1062359011020026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Messina G, Biressi S, Monteverde S, Magli A, Cassano M, Perani L, Roncaglia E, Tagliafico E, Starnes L, Campbell CE, Grossi M, Goldhamer DJ, Gronostajski RM, Cossu G. Nfix regulates fetal-specific transcription in developing skeletal muscle. Cell 2010; 140:554-66. [PMID: 20178747 DOI: 10.1016/j.cell.2010.01.027] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 11/04/2009] [Accepted: 01/14/2010] [Indexed: 12/29/2022]
Abstract
Skeletal myogenesis, like hematopoiesis, occurs in successive developmental stages that involve different cell populations and expression of different genes. We show here that the transcription factor nuclear factor one X (Nfix), whose expression is activated by Pax7 in fetal muscle, in turn activates the transcription of fetal specific genes such as MCK and beta-enolase while repressing embryonic genes such as slow myosin. In the case of the MCK promoter, Nfix forms a complex with PKC theta that binds, phosphorylates, and activates MEF2A. Premature expression of Nfix activates fetal and suppresses embryonic genes in embryonic muscle, whereas muscle-specific ablation of Nfix prevents fetal and maintains embryonic gene expression in the fetus. Therefore, Nfix acts as a transcriptional switch from embryonic to fetal myogenesis.
Collapse
|
18
|
Zhang T, Zaal KJM, Sheridan J, Mehta A, Gundersen GG, Ralston E. Microtubule plus-end binding protein EB1 is necessary for muscle cell differentiation, elongation and fusion. J Cell Sci 2009; 122:1401-9. [PMID: 19366726 DOI: 10.1242/jcs.039255] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
During muscle differentiation, microtubule stability, nucleation and orientation all undergo profound changes, which are simultaneous with and possibly necessary for the elongation and fusion of muscle cells. We do not yet understand these events, but they present similarities with the polarized migration of fibroblasts, in which EB1 is necessary for microtubule stabilization. However, it was recently reported that EB3, not EB1, is involved in muscle cell elongation and fusion, and that neither of these two proteins influences microtubule stabilization. To re-examine the role of EB1, we have generated C2 cell lines permanently expressing EB1-targeted shRNAs. In these lines, EB1 is specifically knocked down by more than 90% before any differentiation-related changes can take place. We find that differentiation (assessed by myogenin expression), elongation and fusion are prevented. In addition, two early events that normally precede differentiation - microtubule stabilization and the accumulation of cadherin and beta-catenin on the plasma membrane - are inhibited. Re-expression of EB1 as EB1-GFP restores all aspects of normal differentiation, whereas overexpression of EB3-GFP restores elongation but not fusion. We conclude that EB1 is necessary for the early stages of muscle differentiation.
Collapse
Affiliation(s)
- Tan Zhang
- Light-imaging Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | |
Collapse
|
19
|
Meng W, Mushika Y, Ichii T, Takeichi M. Anchorage of Microtubule Minus Ends to Adherens Junctions Regulates Epithelial Cell-Cell Contacts. Cell 2008; 135:948-59. [DOI: 10.1016/j.cell.2008.09.040] [Citation(s) in RCA: 283] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Revised: 08/20/2008] [Accepted: 09/17/2008] [Indexed: 10/21/2022]
|
20
|
Tanaka Y, Yamaguchi A, Fujikawa T, Sakuma K, Morita I, Ishii K. Expression of mRNA for specific fibroblast growth factors associates with that of the myogenic markers MyoD and proliferating cell nuclear antigen in regenerating and overloaded rat plantaris muscle. Acta Physiol (Oxf) 2008; 194:149-59. [PMID: 18429950 DOI: 10.1111/j.1748-1716.2008.01866.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM To examine the relations between specific fibroblast growth factors (FGFs) and satellite cell activation during muscle regeneration and hypertrophy in vivo, we measured mRNA expression of FGFs and myogenic markers in rat plantaris muscle after bupivacaine administration and synergist ablation. METHODS mRNA levels for MyoD, myogenin, proliferating cell nuclear antigen (PCNA), p21, M-cadherin, Pax7, FGF-1, FGF-2, FGF-3, FGF-4, FGF-5, FGF-6, FGF-7, FGF-8 and hepatocyte growth factor (HGF) were measured continually for up to 72 h after bupivacaine administration and synergist ablation. FGF-5, FGF-7 and HGF proteins were immunostained at 72 h after bupivacaine administration. RESULTS MyoD and PCNA mRNAs started increasing 24 h after bupivacaine administration. Myogenin, p21, M-cadherin and Pax7 mRNAs started to increase after 48 and 72 h. After synergist ablation, MyoD, PCNA, M-cadherin and Pax7 mRNAs had increased at 24 and 48 h, and myogenin and p21 mRNAs at 12 and 24 h. FGF-1, FGF-7 and HGF mRNAs after the treatments started to increase at the same time as MyoD and PCNA mRNAs. FGF-5 was expressed at the same time as MyoD and PCNA mRNAs after bupivacaine administration but did not after the ablation. FGF-2, FGF-3, FGF-4, FGF-6 and FGF-8 mRNAs were not associated with the expression of the myogenic markers. FGF-7 and HGF proteins were expressed in immature muscle fibre nuclei and the extracellular matrix, but FGF-5 protein was preferentially expressed in extracellular matrix. CONCLUSION These results indicate that FGF-1, FGF-7 and HGF are associated with specific myogenic marker expression during muscle regeneration and hypertrophy.
Collapse
Affiliation(s)
- Y Tanaka
- Laboratory of Human Performance and Fitness, Graduate School of Education, Hokkaido University, Sapporo, Hokkaido, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Hepperger C, Mannes A, Merz J, Peters J, Dietzel S. Three-dimensional positioning of genes in mouse cell nuclei. Chromosoma 2008; 117:535-51. [DOI: 10.1007/s00412-008-0168-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Revised: 05/13/2008] [Accepted: 05/13/2008] [Indexed: 11/24/2022]
|
22
|
Fortier M, Comunale F, Kucharczak J, Blangy A, Charrasse S, Gauthier-Rouvière C. RhoE controls myoblast alignment prior fusion through RhoA and ROCK. Cell Death Differ 2008; 15:1221-31. [PMID: 18369372 DOI: 10.1038/cdd.2008.34] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Differentiation of skeletal myoblasts into multinucleated myotubes is a multi-step process orchestrated by several signaling pathways. The Rho small G protein family plays critical roles both during myogenesis induction and myoblast fusion. We report here that in C2C12 myoblasts, expression of RhoE, an atypical member of this family, increases until the onset of myoblast fusion before resuming its basal level once fusion has occurred. We show that RhoE accumulates in elongated, aligned myoblasts prior to fusion and that its expression is also increased during injury-induced skeletal muscle regeneration. Moreover, although RhoE is not required for myogenesis induction, it is essential for myoblast elongation and alignment before fusion and for M-cadherin expression and accumulation at the cell-cell contact sites. Myoblasts lacking RhoE present with defective p190RhoGAP activation and RhoA inhibition at the onset of myoblast fusion. RhoE interacts also with the RhoA effector Rho-associated kinase (ROCK)I whose activity must be downregulated to allow myoblast fusion. Consistently, we show that pharmacological inactivation of RhoA or ROCK restores myoblast fusion in RhoE-deficient myoblasts. RhoE physiological upregulation before myoblast fusion is responsible for the decrease in RhoA and ROCKI activities, which are required for the fusion process. Therefore, we conclude that RhoE is an essential regulator of myoblast fusion.
Collapse
Affiliation(s)
- M Fortier
- Universités Montpellier 2 et 1, CRBM, CNRS, UMR 5237, IFR 122 1919 Route de Mende, 34293 Montpellier, France
| | | | | | | | | | | |
Collapse
|
23
|
Agarwal N, Hardt T, Brero A, Nowak D, Rothbauer U, Becker A, Leonhardt H, Cardoso MC. MeCP2 interacts with HP1 and modulates its heterochromatin association during myogenic differentiation. Nucleic Acids Res 2007; 35:5402-8. [PMID: 17698499 PMCID: PMC2018631 DOI: 10.1093/nar/gkm599] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
There is increasing evidence of crosstalk between epigenetic modifications such as histone and DNA methylation, recognized by HP1 and methyl CpG-binding proteins, respectively. We have previously shown that the level of methyl CpG-binding proteins increased dramatically during myogenesis leading to large-scale heterochromatin reorganization. In this work, we show that the level of HP1 isoforms did not change significantly throughout myogenic differentiation but their localization did. In particular, HP1γ relocalization to heterochromatin correlated with MeCP2 presence. Using co-immunoprecipitation assays, we found that these heterochromatic factors interact in vivo via the chromo shadow domain of HP1 and the first 55 amino acids of MeCP2. We propose that this dynamic interaction of HP1 and MeCP2 increases their concentration at heterochromatin linking two major gene silencing pathways to stabilize transcriptional repression during differentiation.
Collapse
Affiliation(s)
- Noopur Agarwal
- Max Delbrück Center for Molecular Medicine, 13125 Berlin and Ludwig Maximilians University Munich, Biocenter, Department of Biology, 82152 Planegg-Martinsried, Germany
| | - Tanja Hardt
- Max Delbrück Center for Molecular Medicine, 13125 Berlin and Ludwig Maximilians University Munich, Biocenter, Department of Biology, 82152 Planegg-Martinsried, Germany
| | - Alessandro Brero
- Max Delbrück Center for Molecular Medicine, 13125 Berlin and Ludwig Maximilians University Munich, Biocenter, Department of Biology, 82152 Planegg-Martinsried, Germany
| | - Danny Nowak
- Max Delbrück Center for Molecular Medicine, 13125 Berlin and Ludwig Maximilians University Munich, Biocenter, Department of Biology, 82152 Planegg-Martinsried, Germany
| | - Ulrich Rothbauer
- Max Delbrück Center for Molecular Medicine, 13125 Berlin and Ludwig Maximilians University Munich, Biocenter, Department of Biology, 82152 Planegg-Martinsried, Germany
| | - Annette Becker
- Max Delbrück Center for Molecular Medicine, 13125 Berlin and Ludwig Maximilians University Munich, Biocenter, Department of Biology, 82152 Planegg-Martinsried, Germany
| | - Heinrich Leonhardt
- Max Delbrück Center for Molecular Medicine, 13125 Berlin and Ludwig Maximilians University Munich, Biocenter, Department of Biology, 82152 Planegg-Martinsried, Germany
| | - M. Cristina Cardoso
- Max Delbrück Center for Molecular Medicine, 13125 Berlin and Ludwig Maximilians University Munich, Biocenter, Department of Biology, 82152 Planegg-Martinsried, Germany
- *To whom Correspondence should be addressed: +49 30 94062109+49 30 94063343
| |
Collapse
|
24
|
Dairou J, Pluvinage B, Noiran J, Petit E, Vinh J, Haddad I, Mary J, Dupret JM, Rodrigues-Lima F. Nitration of a critical tyrosine residue in the allosteric inhibitor site of muscle glycogen phosphorylase impairs its catalytic activity. J Mol Biol 2007; 372:1009-1021. [PMID: 17689562 DOI: 10.1016/j.jmb.2007.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Revised: 07/04/2007] [Accepted: 07/05/2007] [Indexed: 10/23/2022]
Abstract
Muscle glycogen phosphorylase (GP) is a key enzyme in glucose metabolism, and its impairment can lead to muscle dysfunction. Tyrosine nitration of glycogen phosphorylase occurs during aging and has been suggested to be involved in progressive loss of muscle performance. Here, we show that GP (in its T and R form) is irreversibly impaired by exposure to peroxynitrite, a biological nitrogen species known to nitrate reactive tyrosine residues, and to be involved in physiological and pathological processes. Kinetic and biochemical analysis indicated that irreversible inactivation of GP by peroxynitrite is due to the fast (k(inact)=3 x 10(4) M(-1) s(-1)) nitration of a unique tyrosine residue of the enzyme. Endogenous GP was tyrosine nitrated and irreversibly inactivated in skeletal muscle cells upon exposure to peroxynitrite, with concomitant impairment of glycogen mobilization. Ligand protection assays and mass spectrometry analysis using purified GP suggested that the peroxynitrite-dependent inactivation of the enzyme could be due to the nitration of Tyr613, a key amino acid of the allosteric inhibitor site of the enzyme. Our findings suggest that GP functions may be regulated by tyrosine nitration.
Collapse
MESH Headings
- Allosteric Regulation
- Animals
- Cells, Cultured
- Dose-Response Relationship, Drug
- Glycogen Phosphorylase, Muscle Form/chemistry
- Glycogen Phosphorylase, Muscle Form/genetics
- Glycogen Phosphorylase, Muscle Form/metabolism
- Mice
- Models, Molecular
- Molsidomine/analogs & derivatives
- Molsidomine/metabolism
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/enzymology
- Nitric Oxide Donors/metabolism
- Peroxynitrous Acid/chemistry
- Peroxynitrous Acid/pharmacology
- Protein Structure, Tertiary
- Tyrosine/chemistry
Collapse
Affiliation(s)
- Julien Dairou
- Laboratoire de Cytophysiologie et Toxicologie Cellulaire (EA 1553), Université Paris Diderot-Paris 7, 75005 Paris, France; UFR de Biochimie, Université Paris Diderot-Paris 7, 75005, Paris, France
| | - Benjamin Pluvinage
- Laboratoire de Cytophysiologie et Toxicologie Cellulaire (EA 1553), Université Paris Diderot-Paris 7, 75005 Paris, France
| | - Joseph Noiran
- UFR de Biochimie, Université Paris Diderot-Paris 7, 75005, Paris, France
| | - Emile Petit
- Laboratoire de Cytophysiologie et Toxicologie Cellulaire (EA 1553), Université Paris Diderot-Paris 7, 75005 Paris, France
| | - Joëlle Vinh
- Laboratoire de Spectrométrie de Masse et Neuroprotéome ESPCI - CNRS UMR 7637, 75005 Paris, France
| | - Iman Haddad
- Laboratoire de Spectrométrie de Masse et Neuroprotéome ESPCI - CNRS UMR 7637, 75005 Paris, France
| | - Jean Mary
- UFR de Biochimie, Université Paris Diderot-Paris 7, 75005, Paris, France; Laboratoire de Biologie et Biochimie Cellulaire du Vieillissement (EA 3106), Université Paris Diderot-Paris 7, 75005 Paris, France
| | - Jean-Marie Dupret
- Laboratoire de Cytophysiologie et Toxicologie Cellulaire (EA 1553), Université Paris Diderot-Paris 7, 75005 Paris, France; UFR de Biochimie, Université Paris Diderot-Paris 7, 75005, Paris, France
| | - Fernando Rodrigues-Lima
- Laboratoire de Cytophysiologie et Toxicologie Cellulaire (EA 1553), Université Paris Diderot-Paris 7, 75005 Paris, France; UFR de Biochimie, Université Paris Diderot-Paris 7, 75005, Paris, France.
| |
Collapse
|
25
|
Charrasse S, Comunale F, Fortier M, Portales-Casamar E, Debant A, Gauthier-Rouvière C. M-cadherin activates Rac1 GTPase through the Rho-GEF trio during myoblast fusion. Mol Biol Cell 2007; 18:1734-43. [PMID: 17332503 PMCID: PMC1855016 DOI: 10.1091/mbc.e06-08-0766] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cadherins are transmembrane glycoproteins that mediate Ca(2+)-dependent homophilic cell-cell adhesion and play crucial role during skeletal myogenesis. M-cadherin is required for myoblast fusion into myotubes, but its mechanisms of action remain unknown. The goal of this study was to cast some light on the nature of the M-cadherin-mediated signals involved in myoblast fusion into myotubes. We found that the Rac1 GTPase activity is increased at the time of myoblast fusion and it is required for this process. Moreover, we showed that M-cadherin-dependent adhesion activates Rac1 and demonstrated the formation of a multiproteic complex containing M-cadherin, the Rho-GEF Trio, and Rac1 at the onset of myoblast fusion. Interestingly, Trio knockdown efficiently blocked both the increase in Rac1-GTP levels, observed after M-cadherin-dependent contact formation, and myoblast fusion. We conclude that M-cadherin-dependent adhesion can activate Rac1 via the Rho-GEF Trio at the time of myoblast fusion.
Collapse
Affiliation(s)
- Sophie Charrasse
- Centre de Recherches de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique, IFR 122, 34293 Montpellier, France
| | - Franck Comunale
- Centre de Recherches de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique, IFR 122, 34293 Montpellier, France
| | - Mathieu Fortier
- Centre de Recherches de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique, IFR 122, 34293 Montpellier, France
| | - Elodie Portales-Casamar
- Centre de Recherches de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique, IFR 122, 34293 Montpellier, France
| | - Anne Debant
- Centre de Recherches de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique, IFR 122, 34293 Montpellier, France
| | - Cécile Gauthier-Rouvière
- Centre de Recherches de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique, IFR 122, 34293 Montpellier, France
| |
Collapse
|
26
|
Wróbel E, Brzóska E, Moraczewski J. M-cadherin and β-catenin participate in differentiation of rat satellite cells. Eur J Cell Biol 2007; 86:99-109. [PMID: 17222478 DOI: 10.1016/j.ejcb.2006.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Revised: 11/09/2006] [Accepted: 11/13/2006] [Indexed: 10/23/2022] Open
Abstract
Cadherins belong to a large family of membrane glycoprotein adhesion receptors that mediate homophilic, calcium-dependent cell adhesion. During myogenesis, cadherins are involved in initial cell-to-cell recognition; and it has also been suggested that they play a role in the initiation of myoblast fusion into multinuclear myotubes. One of the members of the cadherin family, M-cadherin, has been detected during embryogenesis in myogenic cells of somitic origin and in adult muscles. We investigated the distribution and function of M-cadherin and beta-catenin during differentiation of myoblasts in primary cultures of rat satellite cells. We found that M-cadherin was accumulated at the areas of contact between fusing myoblasts and that it colocalized with beta-catenin. Moreover, beta-catenin colocalized with actin in pre-fusing myoblasts. We show that myoblast differentiation is accompanied by an increase in the amounts of M-cadherin and beta-catenin both at the mRNA and the protein level. Flow cytometry analysis showed that M-cadherin expression was highest in fusing myoblasts. In addition, an antibody specific for the extracellular domain of M-cadherin inhibited the fusion of cultured myoblasts. These data suggest that regulation of the M-cadherin level plays an important role in the differentiation of satellite cells and in myoblast fusion in primary cultures.
Collapse
Affiliation(s)
- Edyta Wróbel
- Department of Cytology, Faculty of Biology, Warsaw University, 1 Miecznikowa Street, PL-02-096 Warsaw, Poland
| | | | | |
Collapse
|
27
|
Ishido M, Uda M, Masuhara M, Kami K. Alterations of M-cadherin, neural cell adhesion molecule and beta-catenin expression in satellite cells during overload-induced skeletal muscle hypertrophy. Acta Physiol (Oxf) 2006; 187:407-18. [PMID: 16776666 DOI: 10.1111/j.1748-1716.2006.01577.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM Neural cell adhesion molecule (NCAM) and M-cadherin are cell adhesion molecules expressed on the surface of skeletal muscle satellite cell (SC). During myogenic morphogenesis, M-cadherin participates in mediating terminal differentiation and fusion of myoblasts by forming a complex with beta-catenin and that NCAM contributes to myotube formation by fusion of myoblasts. Hypertrophy and hyperplasia of functionally overloaded skeletal muscle results from the fusion with SCs into the existing myofibres or new myofibre formation by SC-SC fusion. However, the alterations of NCAM, M-cadherin and beta-catenin expressions in SCs in response to functional overload have not been investigated. METHODS Using immunohistochemical approaches, we examined the temporal and spatial expression patterns of these factors expressed in SCs during the functional overload of skeletal muscles. RESULTS Myofibres with SCs showing NCAM+/M-cadherin-, NCAM+/M-cadherin+ or NCAM-/M-cadherin+ were detected in overloaded muscles. The percentage changes of myofibres with SCs showing NCAM+/M-cadherin-, NCAM+/M-cadherin+ or NCAM-/M-cadherin+ were elevated in day-3 post-overloaded muscles, and then only the percentage changes of myofibres with SCs showing NCAM-/M-cadherin+ were significantly increased in day-7 post-overload muscles (P < 0.05). Both beta-catenin and M-cadherin were co-localized throughout quiescent, proliferation and differentiation stages of SCs. CONCLUSION These results suggested that the expressions of NCAM, M-cadherin and beta-catenin in SCs may be controlled by distinct regulatory mechanisms during functional overload, and that interactions among NCAM, M-cadherin and beta-catenin in SCs may play important roles to contribute to overload-induced muscle hypertrophy via fusion with each other or into the existing myofibres of SCs.
Collapse
Affiliation(s)
- M Ishido
- Graduate School of Sport and Exercise Science, Osaka University of Health and Sport Sciences, 1-1 Asashiro-dai, Kumatori-cho, Sennan-gun, Osaka 590-0496, Japan. ,jp
| | | | | | | |
Collapse
|
28
|
Frock RL, Kudlow BA, Evans AM, Jameson SA, Hauschka SD, Kennedy BK. Lamin A/C and emerin are critical for skeletal muscle satellite cell differentiation. Genes Dev 2006; 20:486-500. [PMID: 16481476 PMCID: PMC1369050 DOI: 10.1101/gad.1364906] [Citation(s) in RCA: 204] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Mutations within LMNA, encoding A-type nuclear lamins, are associated with multiple tissue-specific diseases, including Emery-Dreifuss (EDMD2/3) and Limb-Girdle muscular dystrophy (LGMD1B). X-linked EDMD results from mutations in emerin, a lamin A-associated protein. The mechanisms through which these mutations cause muscular dystrophy are not understood. Here we show that most, but not all, cultured muscle cells from lamin A/C knockout mice exhibit impaired differentiation kinetics and reduced differentiation potential. Similarly, normal muscle cells that have been RNA interference (RNAi) down-regulated for either A-type lamins or emerin have impaired differentiation potentials. Replicative myoblasts lacking A-type lamins or emerin also have decreased levels of proteins important for muscle differentiation including pRB, MyoD, desmin, and M-cadherin; up-regulated Myf5; but no changes in Pax3, Pax7, MEF2C, MEF2D, c-met, and beta-catenin. To determine whether impaired myogenesis is linked to reduced MyoD or desmin levels, these proteins were individually expressed in Lmna(-/-) myoblasts that were then induced to undergo myogenesis. Expression of either MyoD or, more surprisingly, desmin in Lmna(-/-) myoblasts resulted in increased differentiation potential. These studies indicate roles for A-type lamins and emerin in myogenic differentiation and also suggest that these effects are at least in part due to decreased endogenous levels of other critical myoblast proteins. The delayed differentiation kinetics and decreased differentiation potential of lamin A/C-deficient and emerin-deficient myoblasts may in part underlie the dystrophic phenotypes observed in patients with EDMD.
Collapse
Affiliation(s)
- Richard L Frock
- Department of Biochemistry, Molecular and Cellular Biology Program, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | |
Collapse
|
29
|
Stehbens SJ, Paterson AD, Crampton MS, Shewan AM, Ferguson C, Akhmanova A, Parton RG, Yap AS. Dynamic microtubules regulate the local concentration of E-cadherin at cell-cell contacts. J Cell Sci 2006; 119:1801-11. [PMID: 16608875 DOI: 10.1242/jcs.02903] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In contrast to the well-established relationship between cadherins and the actin cytoskeleton, the potential link between cadherins and microtubules (MTs) has been less extensively investigated. We now identify a pool of MTs that extend radially into cell-cell contacts and are inhibited by manoeuvres that block the dynamic activity of MT plus-ends (e.g. in the presence of low concentrations of nocodazole and following expression of a CLIP-170 mutant). Blocking dynamic MTs perturbed the ability of cells to concentrate and accumulate E-cadherin at cell-cell contacts, as assessed both by quantitative immunofluorescence microscopy and fluorescence recovery after photobleaching (FRAP) analysis, but did not affect either transport of E-cadherin to the plasma membrane or the amount of E-cadherin expressed at the cell surface. This indicated that dynamic MTs allow cells to concentrate E-cadherin at cell-cell contacts by regulating the regional distribution of E-cadherin once it reaches the cell surface. Importantly, dynamic MTs were necessary for myosin II to accumulate and be activated at cadherin adhesive contacts, a mechanism that supports the focal accumulation of E-cadherin. We propose that this population of MTs represents a novel form of cadherin-MT cooperation, where cadherin adhesions recruit dynamic MTs that, in turn, support the local concentration of cadherin molecules by regulating myosin II activity at cell-cell contacts.
Collapse
Affiliation(s)
- Samantha J Stehbens
- Division of Molecular Cell Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Mayer R, Brero A, von Hase J, Schroeder T, Cremer T, Dietzel S. Common themes and cell type specific variations of higher order chromatin arrangements in the mouse. BMC Cell Biol 2005; 6:44. [PMID: 16336643 PMCID: PMC1325247 DOI: 10.1186/1471-2121-6-44] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2005] [Accepted: 12/07/2005] [Indexed: 11/10/2022] Open
Abstract
Background Similarities as well as differences in higher order chromatin arrangements of human cell types were previously reported. For an evolutionary comparison, we now studied the arrangements of chromosome territories and centromere regions in six mouse cell types (lymphocytes, embryonic stem cells, macrophages, fibroblasts, myoblasts and myotubes) with fluorescence in situ hybridization and confocal laser scanning microscopy. Both species evolved pronounced differences in karyotypes after their last common ancestors lived about 87 million years ago and thus seem particularly suited to elucidate common and cell type specific themes of higher order chromatin arrangements in mammals. Results All mouse cell types showed non-random correlations of radial chromosome territory positions with gene density as well as with chromosome size. The distribution of chromosome territories and pericentromeric heterochromatin changed during differentiation, leading to distinct cell type specific distribution patterns. We exclude a strict dependence of these differences on nuclear shape. Positional differences in mouse cell nuclei were less pronounced compared to human cell nuclei in agreement with smaller differences in chromosome size and gene density. Notably, the position of chromosome territories relative to each other was very variable. Conclusion Chromosome territory arrangements according to chromosome size and gene density provide common, evolutionary conserved themes in both, human and mouse cell types. Our findings are incompatible with a previously reported model of parental genome separation.
Collapse
Affiliation(s)
- Robert Mayer
- Ludwig-Maximilians-Universität München, Department Biologie II, Groβhaderner Str 2, 82152 Planegg-Martinsried, Germany
| | - Alessandro Brero
- Ludwig-Maximilians-Universität München, Department Biologie II, Groβhaderner Str 2, 82152 Planegg-Martinsried, Germany
| | - Johann von Hase
- Kirchhoff Institut für Physik, Universität Heidelberg, Germany
| | - Timm Schroeder
- Institute of Stem Cell Research, GSF – National Research Center for Environment and Health, Neuherberg, Germany
| | - Thomas Cremer
- Ludwig-Maximilians-Universität München, Department Biologie II, Groβhaderner Str 2, 82152 Planegg-Martinsried, Germany
| | - Steffen Dietzel
- Ludwig-Maximilians-Universität München, Department Biologie II, Groβhaderner Str 2, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
31
|
Dairou J, Dupret JM, Rodrigues-Lima F. Impairment of the activity of the xenobiotic-metabolizing enzymes arylamine N-acetyltransferases 1 and 2 (NAT1/NAT2) by peroxynitrite in mouse skeletal muscle cells. FEBS Lett 2005; 579:4719-23. [PMID: 16098511 DOI: 10.1016/j.febslet.2005.07.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2005] [Accepted: 07/21/2005] [Indexed: 11/28/2022]
Abstract
Reactive nitrogen species and their by-products, such as peroxynitrite, modulate many physiological functions of skeletal muscle. Peroxynitrite generation occuring under specific conditions, such as inflammation, may also lead to skeletal muscle dysfunction and pathologies. Arylamine N-acetyltransferases (NATs) are xenobiotic-metabolizing enzymes (XMEs) involved in the detoxification and/or metabolic activation of several drugs and chemicals. In addition to other XMEs, such as gluthatione S-transferases or cytochromes P450, NAT enzymes are expressed in skeletal muscle. We show here that functional NAT1 and NAT2 isoforms are expressed in mouse myotubes and that peroxynitrite may impair their activity in these cells. We show that this inactivation is likely due to the irreversible modification of NATs catalytic cysteine residue in vivo. Our results suggest that peroxynitrite-dependent inactivation of muscle XMEs such as NATs may contribute to muscle dysfunction by impairing the biotransformation activity of this key cellular defense enzyme system.
Collapse
Affiliation(s)
- Julien Dairou
- Laboratoire de Cytophysiologie et Toxicologie Cellulaire, EA 1553, Université Denis Diderot-Paris 7, Case 7073, 75005 Paris, France
| | | | | |
Collapse
|
32
|
Duckmanton A, Kumar A, Chang YT, Brockes JP. A Single-Cell Analysis of Myogenic Dedifferentiation Induced by Small Molecules. ACTA ACUST UNITED AC 2005; 12:1117-26. [PMID: 16242654 DOI: 10.1016/j.chembiol.2005.07.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Revised: 06/01/2005] [Accepted: 07/19/2005] [Indexed: 02/07/2023]
Abstract
An important direction in chemical biology is the derivation of compounds that affect cellular differentiation or its reversal. The fragmentation of multinucleate myofibers into viable mononucleates (called cellularization) occurs during limb regeneration in urodele amphibians, and the isolation of myoseverin, a trisubstituted purine that could apparently activate this pathway of myogenic dedifferentiation in mammalian cells, generated considerable interest. We have explored the mechanism and outcome of cellularization at a single-cell level, and we report findings that significantly extend the previous work with myoseverin. Using a panel of compounds, including a triazine compound with structural similarity and comparable activity to myoseverin, we have identified microtubule disruption as critical for activation of the response. Time-lapse microscopy has enabled us to analyze the fate of identified mononucleate progeny, and directly assess the extent of dedifferentiation.
Collapse
Affiliation(s)
- Amy Duckmanton
- Department of Biochemistry and Molecular Biology, University College London, UK
| | | | | | | |
Collapse
|
33
|
d'Azzo A, Bongiovanni A, Nastasi T. E3 ubiquitin ligases as regulators of membrane protein trafficking and degradation. Traffic 2005; 6:429-41. [PMID: 15882441 DOI: 10.1111/j.1600-0854.2005.00294.x] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ubiquitination is a regulated post-translational modification that conjugates ubiquitin (Ub) to lysine residues of target proteins and determines their intracellular fate. The canonical role of ubiquitination is to mediate degradation by the proteasome of short-lived cytoplasmic proteins that carry a single, polymeric chain of Ub on a specific lysine residue. However, protein modification by Ub has much broader and diverse functions involved in a myriad of cellular processes. Monoubiquitination, at one or multiple lysine residues of transmembrane proteins, influences their stability, protein-protein recognition, activity and intracellular localization. In these processes, Ub functions as an internalization signal that sends the modified substrate to the endocytic/sorting compartments, followed by recycling to the plasma membrane or degradation in the lysosome. E3 ligases play a pivotal role in ubiquitination, because they recognize the acceptor protein and hence dictate the high specificity of the reaction. The multitude of E3s present in nature suggests their nonredundant mode of action and the need for their controlled regulation. Here we give a short account of E3 ligases that specifically modify and regulate membrane proteins. We emphasize the intricate network of interacting proteins that contribute to the substrate-E3 recognition and determine the substrate's cellular fate.
Collapse
Affiliation(s)
- Alessandra d'Azzo
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | | | | |
Collapse
|
34
|
Brero A, Easwaran HP, Nowak D, Grunewald I, Cremer T, Leonhardt H, Cardoso MC. Methyl CpG-binding proteins induce large-scale chromatin reorganization during terminal differentiation. ACTA ACUST UNITED AC 2005; 169:733-43. [PMID: 15939760 PMCID: PMC2171616 DOI: 10.1083/jcb.200502062] [Citation(s) in RCA: 185] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Pericentric heterochromatin plays an important role in epigenetic gene regulation. We show that pericentric heterochromatin aggregates during myogenic differentiation. This clustering leads to the formation of large chromocenters and correlates with increased levels of the methyl CpG–binding protein MeCP2 and pericentric DNA methylation. Ectopic expression of fluorescently tagged MeCP2 mimicked this effect, causing a dose-dependent clustering of chromocenters in the absence of differentiation. MeCP2-induced rearrangement of heterochromatin occurred throughout interphase, did not depend on the H3K9 histone methylation pathway, and required the methyl CpG–binding domain (MBD) only. Similar to MeCP2, another methyl CpG–binding protein, MBD2, also increased during myogenic differentiation and could induce clustering of pericentric regions, arguing for functional redundancy. This MeCP2- and MBD2-mediated chromatin reorganization may thus represent a molecular link between nuclear genome topology and the epigenetic maintenance of cellular differentiation.
Collapse
MESH Headings
- Animals
- Cell Differentiation/genetics
- Cells, Cultured
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/metabolism
- DNA Methylation
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Epigenesis, Genetic/genetics
- Gene Expression Regulation, Developmental/genetics
- Heterochromatin/genetics
- Heterochromatin/metabolism
- Heterochromatin/ultrastructure
- Histones/genetics
- Histones/metabolism
- Male
- Methyl-CpG-Binding Protein 2
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Muscle, Skeletal/embryology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/ultrastructure
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/ultrastructure
- Protein Structure, Tertiary/genetics
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
Collapse
Affiliation(s)
- Alessandro Brero
- Department of Biology II, Ludwig Maximilians University Munich, 82152 Planegg-Martinsried, Germany
| | | | | | | | | | | | | |
Collapse
|
35
|
Tian Q, Feetham MC, Tao WA, He XC, Li L, Aebersold R, Hood L. Proteomic analysis identifies that 14-3-3zeta interacts with beta-catenin and facilitates its activation by Akt. Proc Natl Acad Sci U S A 2004; 101:15370-5. [PMID: 15492215 PMCID: PMC524456 DOI: 10.1073/pnas.0406499101] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
beta-Catenin is a central effector of Wnt signaling in embryonic and stem cell development and in tumorigenesis. Here, through a mass spectrometric analysis of a beta-catenin protein complex, we identified 12 proteins as putative beta-catenin interactors. We show that one of them, 14-3-3zeta, enhances beta-catenin-dependent transcription by maintaining a high level of beta-catenin protein in the cytoplasm. More importantly, 14-3-3zeta facilitates activation of beta-catenin by the survival kinase Akt and colocalizes with activated Akt in intestinal stem cells. We propose that Akt phosphorylates beta-catenin, which results in 14-3-3zeta binding and stabilization of beta-catenin, and these interactions may be involved in stem cell development.
Collapse
Affiliation(s)
- Qiang Tian
- Institute for Systems Biology, Seattle, WA 98103, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Nastasi T, Bongiovanni A, Campos Y, Mann L, Toy JN, Bostrom J, Rottier R, Hahn C, Conaway JW, Harris AJ, D'Azzo A. Ozz-E3, a muscle-specific ubiquitin ligase, regulates beta-catenin degradation during myogenesis. Dev Cell 2004; 6:269-82. [PMID: 14960280 DOI: 10.1016/s1534-5807(04)00020-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2002] [Revised: 04/04/2003] [Accepted: 12/19/2003] [Indexed: 02/06/2023]
Abstract
The identities of the ubiquitin-ligases active during myogenesis are largely unknown. Here we describe a RING-type E3 ligase complex specified by the adaptor protein, Ozz, a novel SOCS protein that is developmentally regulated and expressed exclusively in striated muscle. In mice, the absence of Ozz results in overt maturation defects of the sarcomeric apparatus. We identified beta-catenin as one of the target substrates of the Ozz-E3 in vivo. In the differentiating myofibers, Ozz-E3 regulates the levels of sarcolemma-associated beta-catenin by mediating its degradation via the proteasome. Expression of beta-catenin mutants that reduce the binding of Ozz to endogenous beta-catenin leads to Mb-beta-catenin accumulation and myofibrillogenesis defects similar to those observed in Ozz null myocytes. These findings reveal a novel mechanism of regulation of Mb-beta-catenin and the role of this pool of the protein in myofibrillogenesis, and implicate the Ozz-E3 ligase in the process of myofiber differentiation.
Collapse
MESH Headings
- Aging
- Animals
- Blotting, Northern
- Blotting, Western
- Cells, Cultured
- Cytoskeletal Proteins/metabolism
- Embryo, Mammalian
- Epoxy Compounds/metabolism
- Gene Expression Regulation, Developmental
- Heart
- Humans
- Immunohistochemistry
- In Vitro Techniques
- Mice
- Mice, Knockout
- Microscopy, Electron
- Muscle Development/genetics
- Muscle Development/physiology
- Muscle, Skeletal/abnormalities
- Muscle, Skeletal/embryology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/ultrastructure
- Mutation
- Myoblasts/metabolism
- Myoblasts/ultrastructure
- Myogenin/metabolism
- Phenylalanine/analogs & derivatives
- Phenylalanine/genetics
- Phenylalanine/metabolism
- Precipitin Tests
- Repressor Proteins/metabolism
- Sarcomeres/metabolism
- Sarcomeres/ultrastructure
- Staining and Labeling
- Subcellular Fractions/metabolism
- Tenascin/metabolism
- Trans-Activators/metabolism
- Ubiquitin-Protein Ligase Complexes
- Ubiquitin-Protein Ligases/metabolism
- Ubiquitins/metabolism
- beta Catenin
Collapse
Affiliation(s)
- Tommaso Nastasi
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, 332 North Lauderdale Street, Memphis, TN 38105, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Cortés F, Daggett D, Bryson-Richardson RJ, Neyt C, Maule J, Gautier P, Hollway GE, Keenan D, Currie PD. Cadherin-mediated differential cell adhesion controls slow muscle cell migration in the developing zebrafish myotome. Dev Cell 2004; 5:865-76. [PMID: 14667409 DOI: 10.1016/s1534-5807(03)00362-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Slow-twitch muscle fibers of the zebrafish myotome undergo a unique set of morphogenetic cell movements. During embryogenesis, slow-twitch muscle derives from the adaxial cells, a layer of paraxial mesoderm that differentiates medially within the myotome, immediately adjacent to the notochord. Subsequently, slow-twitch muscle cells migrate through the entire myotome, coming to lie at its most lateral surface. Here we examine the cellular and molecular basis for slow-twitch muscle cell migration. We show that slow-twitch muscle cell morphogenesis is marked by behaviors typical of cells influenced by differential cell adhesion. Dynamic and reciprocal waves of N-cadherin and M-cadherin expression within the myotome, which correlate precisely with cell migration, generate differential adhesive environments that drive slow-twitch muscle cell migration through the myotome. Removing or altering the expression of either protein within the myotome perturbs migration. These results provide a definitive example of homophilic cell adhesion shaping cellular behavior during vertebrate development.
Collapse
Affiliation(s)
- Fernando Cortés
- Comparative and Developmental Genetics Section, MRC Human Genetics Unit, Edinburgh EH4 2XU, Scotland, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Volonte D, Peoples AJ, Galbiati F. Modulation of myoblast fusion by caveolin-3 in dystrophic skeletal muscle cells: implications for Duchenne muscular dystrophy and limb-girdle muscular dystrophy-1C. Mol Biol Cell 2003; 14:4075-88. [PMID: 14517320 PMCID: PMC207001 DOI: 10.1091/mbc.e03-03-0161] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Caveolae are vesicular invaginations of the plasma membrane. Caveolin-3 is the principal structural component of caveolae in skeletal muscle cells in vivo. We have recently generated caveolin-3 transgenic mice and demonstrated that overexpression of wild-type caveolin-3 in skeletal muscle fibers is sufficient to induce a Duchenne-like muscular dystrophy phenotype. In addition, we have shown that caveolin-3 null mice display mild muscle fiber degeneration and T-tubule system abnormalities. These data are consistent with the mild phenotype observed in Limb-girdle muscular dystrophy-1C (LGMD-1C) in humans, characterized by a approximately 95% reduction of caveolin-3 expression. Thus, caveolin-3 transgenic and null mice represent valid mouse models to study Duchenne muscular dystrophy (DMD) and LGMD-1C, respectively, in humans. Here, we derived conditionally immortalized precursor skeletal muscle cells from caveolin-3 transgenic and null mice. We show that overexpression of caveolin-3 inhibits myoblast fusion to multinucleated myotubes and lack of caveolin-3 enhances the fusion process. M-cadherin and microtubules have been proposed to mediate the fusion of myoblasts to myotubes. Interestingly, we show that M-cadherin is downregulated in caveolin-3 transgenic cells and upregulated in caveolin-3 null cells. For the first time, variations of M-cadherin expression have been linked to a muscular dystrophy phenotype. In addition, we demonstrate that microtubules are disorganized in caveolin-3 null myotubes, indicating the importance of the cytoskeleton network in mediating the phenotype observed in these cells. Taken together, these results propose caveolin-3 as a key player in myoblast fusion and suggest that defects of the fusion process may represent additional molecular mechanisms underlying the pathogenesis of DMD and LGMD-1C in humans.
Collapse
Affiliation(s)
- Daniela Volonte
- Department of Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | |
Collapse
|
39
|
Moyer KE, Ehrlich HP. Modulation of human fibroblast gap junction intercellular communication by hyaluronan. J Cell Physiol 2003; 196:165-70. [PMID: 12767052 DOI: 10.1002/jcp.10288] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The composition of the extracellular matrix changes during dermal repair. Initially, hyaluronan (HA) concentration is high, however, by day 3, HA is eliminated. HA optimizes collagen organization within granulation tissue. One possible mechanism of HA modulation of collagen packing is through the promotion of gap junction intercellular communication (GJIC). Gap junctions are gated channels that allow rapid intercellular communication and synchronization of coupled cell activities. The gap junction channel is composed of connexin (Cx) proteins that form a gated channel between coupled cells. HA is reported to enhance Cx43 expression in transformed fibroblasts. GJIC was quantified by the scrape loading technique and reported as a coupling index. The coupling index for human dermal fibroblasts was 4.6 +/- 0.2, while the coupling index for fibroblasts treated with HA more than doubled to 10.6 +/- 0.7. By Western blot analysis no differences were appreciated in the protein levels of Cx43 or beta-catenin, a protein involved in the translocation of Cx to the cell surface. By immuno-histology Cx43 and beta-catenin were evenly distributed throughout the cell in controls, but in cells treated with HA these proteins were co-localized to the cell surface. Coupled fibroblasts are reported to enhance the organization of collagen fibrils. It is proposed that HA increases the accumulation of Cx43 and beta-catenin on the cell surface, leading to greater GJIC and enhanced collagen organization.
Collapse
Affiliation(s)
- K E Moyer
- Division of Plastic Surgery, Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033-0850, USA
| | | |
Collapse
|
40
|
Dedkov EI, Borisov AB, Wernig A, Carlson BM. Aging of skeletal muscle does not affect the response of satellite cells to denervation. J Histochem Cytochem 2003; 51:853-63. [PMID: 12810836 DOI: 10.1177/002215540305100701] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Satellite cells (SCs) are the main source of new fibers in regenerating skeletal muscles and the key contributor to extra nuclei in growing fibers during postnatal development. Aging results in depletion of the SC population and in the reduction of its proliferative activity. Although it has been previously determined that under conditions of massive fiber death in vivo the regenerative potential of SCs is not impaired in old muscle, no studies have yet tested whether advanced age is a factor that may restrain the response of SCs to muscle denervation. The present study is designed to answer this question, comparing the changes of SC numbers in tibialis anterior (TA) muscles from young (4 months) and old (24 months) WI/HicksCar rats after 2 months of denervation. Immunostaining with antibodies against M-cadherin and NCAM was used to detect and count the SCs. The results demonstrate that the percentages of both M-cadherin- and NCAM-positive SCs (SC/Fibers x 100) in control TA muscles from young rats (5.6 +/- 0.5% and 1.4 +/- 0.2%, respectively) are larger than those in old rats (2.3 +/- 0.3% and 0.5 +/- 0.1%, respectively). At the same time, in 2-month denervated TA muscles the percentages of M-cadherin and NCAM positive SC are increased and reach a level that is comparable between young (16.2 +/- 0.9% and 7.5 +/- 0.5%, respectively) and old (15.9 +/- 0.7% and 10.1 +/- 0.5%, respectively) rats. Based on these data, we suggest that aging does not repress the capacity of SC to become activated and grow in the response to muscle denervation.
Collapse
Affiliation(s)
- Eduard I Dedkov
- Department of Cell and Developmental Biology, 4643 Medical Sciences II Building, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | |
Collapse
|
41
|
Clark KA, McElhinny AS, Beckerle MC, Gregorio CC. Striated muscle cytoarchitecture: an intricate web of form and function. Annu Rev Cell Dev Biol 2003; 18:637-706. [PMID: 12142273 DOI: 10.1146/annurev.cellbio.18.012502.105840] [Citation(s) in RCA: 423] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Striated muscle is an intricate, efficient, and precise machine that contains complex interconnected cytoskeletal networks critical for its contractile activity. The individual units of the sarcomere, the basic contractile unit of myofibrils, include the thin, thick, titin, and nebulin filaments. These filament systems have been investigated intensely for some time, but the details of their functions, as well as how they are connected to other cytoskeletal elements, are just beginning to be elucidated. These investigations have advanced significantly in recent years through the identification of novel sarcomeric and sarcomeric-associated proteins and their subsequent functional analyses in model systems. Mutations in these cytoskeletal components account for a large percentage of human myopathies, and thus insight into the normal functions of these proteins has provided a much needed mechanistic understanding of these disorders. In this review, we highlight the components of striated muscle cytoarchitecture with respect to their interactions, dynamics, links to signaling pathways, and functions. The exciting conclusion is that the striated muscle cytoskeleton, an exquisitely tuned, dynamic molecular machine, is capable of responding to subtle changes in cellular physiology.
Collapse
Affiliation(s)
- Kathleen A Clark
- Department of Cell Biology, University of Arizona, Tucson 85724, USA
| | | | | | | |
Collapse
|
42
|
Affiliation(s)
- Zoe Waibler
- Johann Wolfgang Goethe Universität, Institut für Anthropologie und Humangenetik für Biologen, Siesmayerstr. 70, 60323 Frankfurt am Main, Germany
| | | |
Collapse
|
43
|
Martin B, Schneider R, Janetzky S, Waibler Z, Pandur P, Kühl M, Behrens J, von der Mark K, Starzinski-Powitz A, Wixler V. The LIM-only protein FHL2 interacts with beta-catenin and promotes differentiation of mouse myoblasts. J Cell Biol 2002; 159:113-22. [PMID: 12370240 PMCID: PMC2173499 DOI: 10.1083/jcb.200202075] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
FHL2 is a LIM-domain protein expressed in myoblasts but down-regulated in malignant rhabdomyosarcoma cells, suggesting an important role of FHL2 in muscle development. To investigate the importance of FHL2 during myoblast differentiation, we performed a yeast two-hybrid screen using a cDNA library derived from myoblasts induced for differentiation. We identified beta-catenin as a novel interaction partner of FHL2 and confirmed the specificity of association by direct in vitro binding tests and coimmunoprecipitation assays from cell lysates. Deletion analysis of both proteins revealed that the NH2-terminal part of beta-catenin is sufficient for binding in yeast, but addition of the first armadillo repeat is necessary for binding FHL2 in mammalian cells, whereas the presence of all four LIM domains of FHL2 is needed for the interaction. Expression of FHL2 counteracts beta-catenin-mediated activation of a TCF/LEF-dependent reporter gene in a dose-dependent and muscle cell-specific manner. After injection into Xenopus embryos, FHL2 inhibited the beta-catenin-induced axis duplication. C2C12 mouse myoblasts stably expressing FHL2 show increased myogenic differentiation reflected by accelerated myotube formation and expression of muscle-specific proteins. These data imply that FHL2 is a muscle-specific repressor of LEF/TCF target genes and promotes myogenic differentiation by interacting with beta-catenin.
Collapse
Affiliation(s)
- Bernd Martin
- Institut der Anthropologie und Humangenetik für Biologen, Johann-Wolfgang-Goethe-Universität, 60323 Frankfurt, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Langen RCJ, Schols AMWJ, Kelders MCJM, Van Der Velden JLJ, Wouters EFM, Janssen-Heininger YMW. Tumor necrosis factor-alpha inhibits myogenesis through redox-dependent and -independent pathways. Am J Physiol Cell Physiol 2002; 283:C714-21. [PMID: 12176728 DOI: 10.1152/ajpcell.00418.2001] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Muscle wasting accompanies diseases that are associated with chronic elevated levels of circulating inflammatory cytokines and oxidative stress. We previously demonstrated that tumor necrosis factor-alpha (TNF-alpha) inhibits myogenic differentiation via the activation of nuclear factor-kappaB (NF-kappaB). The goal of the present study was to determine whether this process depends on the induction of oxidative stress. We demonstrate here that TNF-alpha causes a decrease in reduced glutathione (GSH) during myogenic differentiation of C(2)C(12) cells, which coincides with an elevated generation of reactive oxygen species. Supplementation of cellular GSH with N-acetyl-l-cysteine (NAC) did not reverse the inhibitory effects of TNF-alpha on troponin I promoter activation and only partially restored creatine kinase activity in TNF-alpha-treated cells. In contrast, the administration of NAC before treatment with TNF-alpha almost completely restored the formation of multinucleated myotubes. NAC decreased TNF-alpha-induced activation of NF-kappaB only marginally, indicating that the redox-sensitive component of the inhibition of myogenic differentiation by TNF-alpha occurred independently, or downstream of NF-kappaB. Our observations suggest that the inhibitory effects of TNF-alpha on myogenesis can be uncoupled in a redox-sensitive component affecting myotube formation and a redox independent component affecting myogenic protein expression.
Collapse
Affiliation(s)
- Ramon C J Langen
- Department of Pulmonology, Maastricht University, The Netherlands
| | | | | | | | | | | |
Collapse
|
45
|
Hollnagel A, Grund C, Franke WW, Arnold HH. The cell adhesion molecule M-cadherin is not essential for muscle development and regeneration. Mol Cell Biol 2002; 22:4760-70. [PMID: 12052883 PMCID: PMC133893 DOI: 10.1128/mcb.22.13.4760-4770.2002] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
M-cadherin is a classical calcium-dependent cell adhesion molecule that is highly expressed in developing skeletal muscle, satellite cells, and cerebellum. Based on its expression pattern and observations in cell culture, it has been postulated that M-cadherin may be important for the fusion of myoblasts to form myotubes, the correct localization and function of satellite cells during muscle regeneration, and the specialized architecture of adhering junctions in granule cells of cerebellar glomeruli. In order to investigate the potential roles of M-cadherin in vivo, we generated a null mutation in mice. Mutant mice were viable and fertile and showed no gross developmental defects. In particular, the skeletal musculature appeared essentially normal. Moreover, muscle lesions induced by necrosis were efficiently repaired in mutant mice, suggesting that satellite cells are present, can be activated, and are able to form new myofibers. This was also confirmed by normal growth and fusion potential of mutant satellite cells cultured in vitro. In the cerebellum of M-cadherin-lacking mutants, typical contactus adherens junctions were present and similar in size and numbers to the equivalent junctions in wild-type animals. However, the adhesion plaques in the cerebellum of these mutants appeared to contain elevated levels of N-cadherin compared to wild-type animals. Taken together, these observations suggest that M-cadherin in the mouse serves no absolutely required function during muscle development and regeneration and is not essential for the formation of specialized cell contacts in the cerebellum. It seems that N-cadherin or other cadherins can largely compensate for the lack of M-cadherin.
Collapse
Affiliation(s)
- Angela Hollnagel
- Department of Cell and Molecular Biology, Institute of Biochemistry and Biotechnology, Technical University of Braunschweig, 38106 Braunschweig, Germany
| | | | | | | |
Collapse
|
46
|
Waibler Z, Schäfer A, Starzinski-Powitz A. mARVCF cellular localisation and binding to cadherins is influenced by the cellular context but not by alternative splicing. J Cell Sci 2001; 114:3873-84. [PMID: 11719554 DOI: 10.1242/jcs.114.21.3873] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ARVCF, a member of the catenin family, is thought to contribute to the morphoregulatory function of the cadherin-catenin complex. Recently, we reported the isolation and characterisation of murine ARVCF (mARVCF), particularly its interaction with M-cadherin. Here, we describe the identification of novel mARVCF isoforms that arise by alternative splicing. At the N-terminus, alternative splicing results in the inclusion or omission of a coiled-coil region probably important for protein-protein interactions. At the C-terminus, four isoforms also differ by domains potentially important for selective protein-protein interaction. The eight putative mARVCF isoforms were expressed as EGFP-fusion proteins in six different cell lines that exhibit a distinct pattern of cadherins. Apparently, binding of the mARVCF isoforms to M-, N-, or E-cadherin is generally unaffected by their altered N- and C-termini, as revealed by the MOM recruitment assay. However, mARVCF isoforms reproducibly exhibit differential localisation in distinct cellular environments. For example, mARVCF isoforms are unable to colocalise with N-cadherin in EJ28 carcinoma cells but do so in HeLa cells. Our results suggest that the subcellular localisation of mARVCF may be determined not only by the presence or absence of an appropriate interaction partner, in this case cadherins, but also by the cellular context.
Collapse
Affiliation(s)
- Z Waibler
- Institut der Anthropologie und Humangenetik fuer Biologen, Johann-Wolfgang-Goethe-Universitaet Frankfurt, Siesmayerstrasse 70, D-60054 Frankfurt/Main, Germany
| | | | | |
Collapse
|
47
|
Link D, Irintchev A, Knauf U, Wernig A, Starzinski-Powitz A. A model system for studying postnatal myogenesis with tetracycline-responsive, genetically engineered clonal myoblasts in vitro and in vivo. Exp Cell Res 2001; 270:138-50. [PMID: 11640878 DOI: 10.1006/excr.2001.5340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this work was to introduce a tetracycline-responsive (Tet-off) gene expression system into myoblasts in order to regulate a reporter gene not only in vitro but also particularly in muscles implanted with these engineered myoblasts. Mouse myoblasts from a long-term culture (i28 cells) were transfected initially to generate and characterize two stable master clones expressing tetracycline-responsive transactivator protein tTA. Like parental i28 myoblasts, these clones differentiated well in vitro. The second step introduced the firefly (Photinus pyralis) luciferase gene into one of the stable tTA clones producing double transfectants expressing luciferase in the absence of tetracycline. Addition of tetracycline (1 microg ml(-1)) resulted in at least 100-fold decreases in luciferase activity within 8 h in both growing and differentiating myoblast cultures. Enzyme activity was rapidly restored after tetracycline was removed (8 h). After successful implantation of these myoblasts into damaged mouse muscles, luciferase expression in the matured progeny cells could be regulated by oral application of doxycycline for at least 1 month. The tetracycline-responsive master clones are potentially powerful tools for studying the function of various genes in postnatal myogenesis.
Collapse
Affiliation(s)
- D Link
- Xantos Biomedicine AG, Fraunhoferstrasse 22, Martinsried, D-82152, Germany
| | | | | | | | | |
Collapse
|
48
|
Kee SH, Steinert PM. Microtubule disruption in keratinocytes induces cell-cell adhesion through activation of endogenous E-cadherin. Mol Biol Cell 2001; 12:1983-93. [PMID: 11451997 PMCID: PMC55644 DOI: 10.1091/mbc.12.7.1983] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The association of the cytoskeleton with the cadherin--catenin complex is essential for strong cell-cell adhesion in epithelial cells. In this study, we have investigated the effect of microtubule organization on cell-cell adhesion in differentiating keratinocytes. When microtubules of normal human epidermal keratinocytes (NHEKs) grown in low calcium media (0.05 mM) were disrupted with nocodazole or colcemid, cell-cell adhesion was induced through relocalization of the E-cadherin-catenin-actin complex to the cell periphery. This was accompanied by actin polymerization. Also, it was found that microtubule disruption-induced cell-cell adhesion was significantly reduced in more advanced differentiated keratinocytes. For example, when NHEK cells cultured under high calcium (1.2 mM) for 8 d and then in low calcium for 1 d were treated with nocodazole, there was no induction of cell-cell adhesion. Also long-term treatment of a phorbol ester for 48 h inhibited nocodazole-induced cell-cell adhesion of NHEK. Furthermore, this nocodazole-induced cell-cell adhesion could be observed in squamous cancer cell lines (A431 and SCC-5, -9, and -25) under low calcium condition, but not in the keratinocyte cell lines derived from normal epidermis (HaCaT, RHEK). On the other hand, HaCaT cells continuously cultivated in low calcium media regained a less differentiated phenotype such as decreased expression of cytokeratin 10, and increased K5; these changes were accompanied with inducibility of cell-cell adhesion by nocodazole. Together, our results suggest that microtubule disruption can induce the cell-cell adhesion via activation of endogenous E-cadherin in non- or early differentiating keratinocytes. However, this is no longer possible in advanced terminally differentiating keratinocytes, possibly due to irreversible changes effected by cell envelope barrier formation.
Collapse
Affiliation(s)
- S H Kee
- Laboratory of Skin Biology, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892-2752, USA
| | | |
Collapse
|
49
|
Chausovsky A, Bershadsky AD, Borisy GG. Cadherin-mediated regulation of microtubule dynamics. Nat Cell Biol 2000; 2:797-804. [PMID: 11056534 DOI: 10.1038/35041037] [Citation(s) in RCA: 116] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Epithelial polarization and neuronal outgrowth require the assembly of microtubule arrays that are not associated with centrosomes. As these processes generally involve contact interactions mediated by cadherins, we investigated the potential role of cadherin signalling in the stabilization of non-centrosomal microtubules. Here we show that expression of cadherins in centrosome-free cytoplasts increases levels of microtubule polymer and changes the behaviour of microtubules from treadmilling to dynamic instability. This effect is not a result of cadherin expression per se but depends on the formation of cell-cell contacts. The effect of cell-cell contacts is mimicked by application of beads coated with stimulatory anti-cadherin antibody and is suppressed by overexpression of the cytoplasmic cadherin tail. We therefore propose that cadherins initiate a signalling pathway that alters microtubule organization by stabilizing microtubule ends.
Collapse
Affiliation(s)
- A Chausovsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | | | | |
Collapse
|
50
|
Spencer JA, Eliazer S, Ilaria RL, Richardson JA, Olson EN. Regulation of microtubule dynamics and myogenic differentiation by MURF, a striated muscle RING-finger protein. J Cell Biol 2000; 150:771-84. [PMID: 10953002 PMCID: PMC2175279 DOI: 10.1083/jcb.150.4.771] [Citation(s) in RCA: 144] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The RING-finger domain is a novel zinc-binding Cys-His protein motif found in a growing number of proteins involved in signal transduction, ubiquitination, gene transcription, differentiation, and morphogenesis. We describe a novel muscle-specific RING-finger protein (MURF) expressed specifically in cardiac and skeletal muscle cells throughout pre- and postnatal mouse development. MURF belongs to the RING-B-box-coiled-coil subclass of RING-finger proteins, characterized by an NH(2)-terminal RING-finger followed by a zinc-finger domain (B-box) and a leucine-rich coiled-coil domain. Expression of MURF is required for skeletal myoblast differentiation and myotube fusion. The leucine-rich coiled-coil domain of MURF mediates association with microtubules, whereas the RING-finger domain is required for microtubule stabilization and an additional region is required for homo-oligomerization. Expression of MURF establishes a cellular microtubule network that is resistant to microtubule depolymerization induced by alkaloids, cold and calcium. These results identify MURF as a myogenic regulator of the microtubule network of striated muscle cells and reveal a link between microtubule organization and myogenesis.
Collapse
Affiliation(s)
- Jeffrey A. Spencer
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75235-9148
| | - Susan Eliazer
- Department of Hematology and Oncology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75235-9148
| | - Robert L. Ilaria
- Department of Hematology and Oncology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75235-9148
| | - James A. Richardson
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75235-9148
| | - Eric N. Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75235-9148
| |
Collapse
|