1
|
Lohanadan K, Assent M, Linnemann A, Schuld J, Heukamp LC, Krause K, Vorgerd M, Reimann J, Schänzer A, Kirfel G, Fürst DO, Van der Ven PFM. Synaptopodin-2 Isoforms Have Specific Binding Partners and Display Distinct, Muscle Cell Type-Specific Expression Patterns. Cells 2023; 13:85. [PMID: 38201288 PMCID: PMC10778272 DOI: 10.3390/cells13010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Synaptopodin-2 (SYNPO2) is a protein associated with the Z-disc in striated muscle cells. It interacts with α-actinin and filamin C, playing a role in Z-disc maintenance under stress by chaperone-assisted selective autophagy (CASA). In smooth muscle cells, SYNPO2 is a component of dense bodies. Furthermore, it has been proposed to play a role in tumor cell proliferation and metastasis in many different kinds of cancers. Alternative transcription start sites and alternative splicing predict the expression of six putative SYNPO2 isoforms differing by extended amino- and/or carboxy-termini. Our analyses at mRNA and protein levels revealed differential expression of SYNPO2 isoforms in cardiac, skeletal and smooth muscle cells. We identified synemin, an intermediate filament protein, as a novel binding partner of the PDZ-domain in the amino-terminal extension of the isoforms mainly expressed in cardiac and smooth muscle cells, and demonstrated colocalization of SYNPO2 and synemin in both cell types. A carboxy-terminal extension, mainly expressed in smooth muscle cells, is sufficient for association with dense bodies and interacts with α-actinin. SYNPO2 therefore represents an additional and novel link between intermediate filaments and the Z-discs in cardiomyocytes and dense bodies in smooth muscle cells, respectively. In pathological skeletal muscle samples, we identified SYNPO2 in the central and intermediate zones of target fibers of patients with neurogenic muscular atrophy, and in nemaline bodies. Our findings help to understand distinct functions of individual SYNPO2 isoforms in different muscle tissues, but also in tumor pathology.
Collapse
Affiliation(s)
| | - Marvin Assent
- Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Anja Linnemann
- Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Julia Schuld
- Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Lukas C. Heukamp
- Department of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Karsten Krause
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Jens Reimann
- Department of Neurology, Neuromuscular Diseases Section, University Hospital Bonn, 53127 Bonn, Germany
| | - Anne Schänzer
- Institute of Neuropathology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Gregor Kirfel
- Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Dieter O. Fürst
- Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | | |
Collapse
|
2
|
Garcia-Pelagio KP, Bloch RJ. Biomechanical Properties of the Sarcolemma and Costameres of Skeletal Muscle Lacking Desmin. Front Physiol 2021; 12:706806. [PMID: 34489727 PMCID: PMC8416993 DOI: 10.3389/fphys.2021.706806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/13/2021] [Indexed: 01/23/2023] Open
Abstract
Intermediate filaments (IFs), composed primarily by desmin and keratins, link the myofibrils to each other, to intracellular organelles, and to the sarcolemma. There they may play an important role in transfer of contractile force from the Z-disks and M-lines of neighboring myofibrils to costameres at the membrane, across the membrane to the extracellular matrix, and ultimately to the tendon (“lateral force transmission”). We measured the elasticity of the sarcolemma and the connections it makes at costameres with the underlying contractile apparatus of individual fast twitch muscle fibers of desmin-null mice. By positioning a suction pipet to the surface of the sarcolemma and applying increasing pressure, we determined the pressure at which the sarcolemma separated from nearby sarcomeres, Pseparation, and the pressure at which the isolated sarcolemma burst, Pbursting. We also examined the time required for the intact sarcolemma-costamere-sarcomere complex to reach equilibrium at lower pressures. All measurements showed the desmin-null fibers to have slower equilibrium times and lower Pseparation and Pbursting than controls, suggesting that the sarcolemma and its costameric links to nearby contractile structures were weaker in the absence of desmin. Comparisons to earlier values determined for muscles lacking dystrophin or synemin suggest that the desmin-null phenotype is more stable than the former and less stable than the latter. Our results are consistent with the moderate myopathy seen in desmin-null muscles and support the idea that desmin contributes significantly to sarcolemmal stability and lateral force transmission.
Collapse
Affiliation(s)
- Karla P Garcia-Pelagio
- Departamento de Fisica, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Robert J Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
3
|
Potokar M, Morita M, Wiche G, Jorgačevski J. The Diversity of Intermediate Filaments in Astrocytes. Cells 2020; 9:E1604. [PMID: 32630739 PMCID: PMC7408014 DOI: 10.3390/cells9071604] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/26/2020] [Accepted: 07/01/2020] [Indexed: 01/02/2023] Open
Abstract
Despite the remarkable complexity of the individual neuron and of neuronal circuits, it has been clear for quite a while that, in order to understand the functioning of the brain, the contribution of other cell types in the brain have to be accounted for. Among glial cells, astrocytes have multiple roles in orchestrating neuronal functions. Their communication with neurons by exchanging signaling molecules and removing molecules from extracellular space takes place at several levels and is governed by different cellular processes, supported by multiple cellular structures, including the cytoskeleton. Intermediate filaments in astrocytes are emerging as important integrators of cellular processes. Astrocytes express five types of intermediate filaments: glial fibrillary acidic protein (GFAP); vimentin; nestin; synemin; lamins. Variability, interactions with different cellular structures and the particular roles of individual intermediate filaments in astrocytes have been studied extensively in the case of GFAP and vimentin, but far less attention has been given to nestin, synemin and lamins. Similarly, the interplay between different types of cytoskeleton and the interaction between the cytoskeleton and membranous structures, which is mediated by cytolinker proteins, are understudied in astrocytes. The present review summarizes the basic properties of astrocytic intermediate filaments and of other cytoskeletal macromolecules, such as cytolinker proteins, and describes the current knowledge of their roles in normal physiological and pathological conditions.
Collapse
Affiliation(s)
- Maja Potokar
- Laboratory of Neuroendocrinology – Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
- Celica BIOMEDICAL, 1000 Ljubljana, Slovenia;
| | - Mitsuhiro Morita
- Department of Biology, Kobe University Graduate School of Science, Kobe 657-8501, Japan;
| | - Gerhard Wiche
- Celica BIOMEDICAL, 1000 Ljubljana, Slovenia;
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, 1030 Vienna, Austria
| | - Jernej Jorgačevski
- Laboratory of Neuroendocrinology – Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
- Celica BIOMEDICAL, 1000 Ljubljana, Slovenia;
| |
Collapse
|
4
|
Russell MA. Synemin Redefined: Multiple Binding Partners Results in Multifunctionality. Front Cell Dev Biol 2020; 8:159. [PMID: 32258037 PMCID: PMC7090255 DOI: 10.3389/fcell.2020.00159] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022] Open
Abstract
Historically synemin has been studied as an intermediate filament protein. However, synemin also binds the type II regulatory (R) subunit α of protein kinase A (PKA) and protein phosphatase type 2A, thus participating in the PKA and phosphoinositide 3-kinase (PI3K)-Akt and signaling pathways. In addition, recent studies using transgenic mice indicate that a significant function of synemin is its role in signaling pathways in various tissues, including the heart. Recent clinical reports have shown that synemin mutations led to multiple cases of dilated cardiomyopathy. Additionally, a single case of the rare condition ulnar-mammary-like syndrome with left ventricular tachycardia due to a mutation in the synemin gene (SYNM) has been reported. Therefore, this review uses these recent studies to provide a new framework for detailed discussions on synemin tissue distribution, binding partners and synemin in disease. Differences between α- and β-synemin are highlighted. The studies presented here indicate that while synemin does function as an intermediate filament protein, it is unique among this large family of proteins as it is also a regulator of signaling pathways and a crosslinker. Also evident is that the dominant function(s) are isoform-, developmental-, and tissue-specific.
Collapse
Affiliation(s)
- Mary A Russell
- Department of Biological Sciences, Kent State University at Trumbull, Warren, OH, United States
| |
Collapse
|
5
|
Paulin D, Hovhannisyan Y, Kasakyan S, Agbulut O, Li Z, Xue Z. Synemin-related skeletal and cardiac myopathies: an overview of pathogenic variants. Am J Physiol Cell Physiol 2020; 318:C709-C718. [PMID: 32023076 DOI: 10.1152/ajpcell.00485.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review analyzes data concerning patients with cardiomyopathies or skeletal myopathies associated with a variation in the intermediate filament (IF) synemin gene (SYNM), also referred to as desmuslin (DMN). Molecular studies demonstrate that synemin copolymerizes with desmin and vimentin IF and interacts with vinculin, α-actinin, α-dystrobrevin, dystrophin, talin, and zyxin. It has been found that synemin is an A-kinase-anchoring protein (AKAP) that anchors protein kinase A (PKA) and modulates the PKA-dependent phosphorylation of several cytoskeletal substrates such as desmin. Because several IF proteins, including desmin, have been implicated in human genetic disorders such as dominant or recessive congenital and adult-onset myopathy, synemin becomes a significant candidate for cardiac and skeletal myopathies of unknown etiology. Because SYNM is a new candidate gene that displays numerous sequence polymorphisms, in this review, we summarize the genetic and clinical literature about SYNM mutations. Protein-changing variants (missense, frameshifts, nonsense) were further evaluated based on structural modifications and amino acid interactions. We present in silico modeling of helical salt-bridges between residues to evaluate the impact of the synemin networks crucial to interactions with cytoskeletal proteins. Finally, a discussion is featured regarding certain variants that may contribute to the disease state.
Collapse
Affiliation(s)
- Denise Paulin
- Sorbonne Université, Institut de Biologie Paris-Seine, CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Yeranuhi Hovhannisyan
- Sorbonne Université, Institut de Biologie Paris-Seine, CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Serdar Kasakyan
- Duzen Laboratories Group, Center of Genetic Diagnosis, Istanbul, Turkey
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine, CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Zhenlin Li
- Sorbonne Université, Institut de Biologie Paris-Seine, CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Zhigang Xue
- Sorbonne Université, Institut de Biologie Paris-Seine, CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| |
Collapse
|
6
|
Klymkowsky MW. Filaments and phenotypes: cellular roles and orphan effects associated with mutations in cytoplasmic intermediate filament proteins. F1000Res 2019; 8. [PMID: 31602295 PMCID: PMC6774051 DOI: 10.12688/f1000research.19950.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2019] [Indexed: 12/11/2022] Open
Abstract
Cytoplasmic intermediate filaments (IFs) surround the nucleus and are often anchored at membrane sites to form effectively transcellular networks. Mutations in IF proteins (IFps) have revealed mechanical roles in epidermis, muscle, liver, and neurons. At the same time, there have been phenotypic surprises, illustrated by the ability to generate viable and fertile mice null for a number of IFp-encoding genes, including vimentin. Yet in humans, the vimentin ( VIM) gene displays a high probability of intolerance to loss-of-function mutations, indicating an essential role. A number of subtle and not so subtle IF-associated phenotypes have been identified, often linked to mechanical or metabolic stresses, some of which have been found to be ameliorated by the over-expression of molecular chaperones, suggesting that such phenotypes arise from what might be termed "orphan" effects as opposed to the absence of the IF network per se, an idea originally suggested by Toivola et al. and Pekny and Lane.
Collapse
Affiliation(s)
- Michael W Klymkowsky
- Molecular, Cellular & Developmental Biology, University of Colorado, Boulder, Boulder, CO, 80303, USA
| |
Collapse
|
7
|
Swärd K, Krawczyk KK, Morén B, Zhu B, Matic L, Holmberg J, Hedin U, Uvelius B, Stenkula K, Rippe C. Identification of the intermediate filament protein synemin/SYNM as a target of myocardin family coactivators. Am J Physiol Cell Physiol 2019; 317:C1128-C1142. [PMID: 31461342 DOI: 10.1152/ajpcell.00047.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Myocardin (MYOCD) is a critical regulator of smooth muscle cell (SMC) differentiation, but its transcriptional targets remain to be exhaustively characterized, especially at the protein level. Here we leveraged human RNA and protein expression data to identify novel potential MYOCD targets. Using correlation analyses we found several targets that we could confirm at the protein level, including SORBS1, SLMAP, SYNM, and MCAM. We focused on SYNM, which encodes the intermediate filament protein synemin. SYNM rivalled smooth muscle myosin (MYH11) for SMC specificity and was controlled at the mRNA and protein levels by all myocardin-related transcription factors (MRTFs: MYOCD, MRTF-A/MKL1, and MRTF-B/MKL2). MRTF activity is regulated by the ratio of filamentous to globular actin, and SYNM was accordingly reduced by interventions that depolymerize actin, such as latrunculin treatment and overexpression of constitutively active cofilin. Many MRTF target genes depend on serum response factor (SRF), but SYNM lacked SRF-binding motifs in its proximal promoter, which was not directly regulated by MYOCD. Furthermore, SYNM resisted SRF silencing, yet the time course of induction closely paralleled that of the SRF-dependent target gene ACTA2. SYNM was repressed by the ternary complex factor (TCF) FLI1 and was increased in mouse embryonic fibroblasts lacking three classical TCFs (ELK1, ELK3, and ELK4). Imaging showed colocalization of SYNM with the intermediate filament proteins desmin and vimentin, and MRTF-A/MKL1 increased SYNM-containing intermediate filaments in SMCs. These studies identify SYNM as a novel SRF-independent target of myocardin that is abundantly expressed in all SMCs.
Collapse
Affiliation(s)
- Karl Swärd
- Department of Experimental Medical Science, Lund, Sweden
| | | | - Björn Morén
- Department of Experimental Medical Science, Lund, Sweden
| | - Baoyi Zhu
- Department of Experimental Medical Science, Lund, Sweden.,Department of Urology, the Sixth Affiliated Hospital of Guangzhou Medical University (Qingyuan People's Hospital), Guangdong, China
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Johan Holmberg
- Department of Experimental Medical Science, Lund, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Uvelius
- Department of Clinical Science, Lund, Lund University, Lund, Sweden
| | - Karin Stenkula
- Department of Experimental Medical Science, Lund, Sweden
| | - Catarina Rippe
- Department of Experimental Medical Science, Lund, Sweden
| |
Collapse
|
8
|
Rodríguez MA, Liu JX, Parkkonen K, Li Z, Pedrosa Domellöf F. The Cytoskeleton in the Extraocular Muscles of Desmin Knockout Mice. Invest Ophthalmol Vis Sci 2019; 59:4847-4855. [PMID: 30347079 DOI: 10.1167/iovs.18-24508] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the effect of absence of desmin on the extraocular muscles (EOMs) with focus on the structure and composition of the cytoskeleton. Methods The distribution of synemin, syncoilin, plectin, nestin, and dystrophin was evaluated on cross and longitudinal sections of EOMs and limb muscles from 1-year-old desmin knockout mice (desmin-/-) by immunofluorescence. General morphology was evaluated with hematoxylin and eosin while mitochondrial content and distribution were evaluated by succinate dehydrogenase (SDH) and modified Gomori trichrome stainings. Results The muscle fibers of the EOMs in desmin-/- mice were remarkably well preserved in contrast to those in the severely affected soleus and the slightly affected gastrocnemius muscles. There were no signs of muscular pathology in the EOMs and all cytoskeletal proteins studied showed a correct location at sarcolemma and Z-discs. However, an increase of SDH staining and mitochondrial aggregates under the sarcolemma was detected. Conclusions The structure of the EOMs was well preserved in the absence of desmin. We suggest that desmin is not necessary for correct synemin, syncoilin, plectin, and dystrophin location on the cytoskeleton of EOMs. However, it is needed to maintain an appropriate mitochondrial distribution in both EOMs and limb muscles.
Collapse
Affiliation(s)
| | - Jing-Xia Liu
- Department of Integrative Medical Biology, Section for Anatomy, Umeå University, Umeå, Sweden
| | - Kimmo Parkkonen
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden.,Department of Integrative Medical Biology, Section for Anatomy, Umeå University, Umeå, Sweden
| | | | | |
Collapse
|
9
|
The Cytoskeleton-A Complex Interacting Meshwork. Cells 2019; 8:cells8040362. [PMID: 31003495 PMCID: PMC6523135 DOI: 10.3390/cells8040362] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022] Open
Abstract
The cytoskeleton of animal cells is one of the most complicated and functionally versatile structures, involved in processes such as endocytosis, cell division, intra-cellular transport, motility, force transmission, reaction to external forces, adhesion and preservation, and adaptation of cell shape. These functions are mediated by three classical cytoskeletal filament types, as follows: Actin, microtubules, and intermediate filaments. The named filaments form a network that is highly structured and dynamic, responding to external and internal cues with a quick reorganization that is orchestrated on the time scale of minutes and has to be tightly regulated. Especially in brain tumors, the cytoskeleton plays an important role in spreading and migration of tumor cells. As the cytoskeletal organization and regulation is complex and many-faceted, this review aims to summarize the findings about cytoskeletal filament types, including substructures formed by them, such as lamellipodia, stress fibers, and interactions between intermediate filaments, microtubules and actin. Additionally, crucial regulatory aspects of the cytoskeletal filaments and the formed substructures are discussed and integrated into the concepts of cell motility. Even though little is known about the impact of cytoskeletal alterations on the progress of glioma, a final point discussed will be the impact of established cytoskeletal alterations in the cellular behavior and invasion of glioma.
Collapse
|
10
|
Brodehl A, Gaertner-Rommel A, Milting H. Molecular insights into cardiomyopathies associated with desmin (DES) mutations. Biophys Rev 2018; 10:983-1006. [PMID: 29926427 DOI: 10.1007/s12551-018-0429-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/22/2018] [Indexed: 12/15/2022] Open
Abstract
Increasing usage of next-generation sequencing techniques pushed during the last decade cardiogenetic diagnostics leading to the identification of a huge number of genetic variants in about 170 genes associated with cardiomyopathies, channelopathies, or syndromes with cardiac involvement. Because of the biochemical and cellular complexity, it is challenging to understand the clinical meaning or even the relevant pathomechanisms of the majority of genetic sequence variants. However, detailed knowledge about the associated molecular pathomechanism is essential for the development of efficient therapeutic strategies in future and genetic counseling. Mutations in DES, encoding the muscle-specific intermediate filament protein desmin, have been identified in different kinds of cardiac and skeletal myopathies. Here, we review the functions of desmin in health and disease with a focus on cardiomyopathies. In addition, we will summarize the genetic and clinical literature about DES mutations and will explain relevant cell and animal models. Moreover, we discuss upcoming perspectives and consequences of novel experimental approaches like genome editing technology, which might open a novel research field contributing to the development of efficient and mutation-specific treatment options.
Collapse
Affiliation(s)
- Andreas Brodehl
- Erich and Hanna Klessmann Institute for Cardiovascular Research & Development, Heart and Diabetes Centre NRW, Ruhr-University Bochum, Georgstrasse 11, 32545, Bad Oeynhausen, Germany.
| | - Anna Gaertner-Rommel
- Erich and Hanna Klessmann Institute for Cardiovascular Research & Development, Heart and Diabetes Centre NRW, Ruhr-University Bochum, Georgstrasse 11, 32545, Bad Oeynhausen, Germany
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute for Cardiovascular Research & Development, Heart and Diabetes Centre NRW, Ruhr-University Bochum, Georgstrasse 11, 32545, Bad Oeynhausen, Germany.
| |
Collapse
|
11
|
Zlotina A, Kiselev A, Sergushichev A, Parmon E, Kostareva A. Rare Case of Ulnar-Mammary-Like Syndrome With Left Ventricular Tachycardia and Lack of TBX3 Mutation. Front Genet 2018; 9:209. [PMID: 29963074 PMCID: PMC6013977 DOI: 10.3389/fgene.2018.00209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 05/25/2018] [Indexed: 12/13/2022] Open
Abstract
"Heart-hand" type syndromes represent a group of rare congenital conditions that combine cardiac pathology (structural defect or arrhythmic disorder) and limb abnormality. Significant clinical variability and genetic heterogeneity typical for such syndromes complicate correct diagnosis, prognosis, and appropriate genetic counseling of the affected families. By now, only single genes have been unambiguously determined as a genetic cause of heart-hand syndromes and phenotypically similar conditions. In the present study, we report on a 25-year-old Russian female patient with a clinical picture resembling ulnar-mammary syndrome (UMS). Principal clinical manifestations included heart septal fibrosis and non-sustained left ventricular tachycardia combined with fifth finger camptodactyly, hypoplastic breast, abnormal teeth, and mental retardation. Target Sanger sequencing and array-based comparative genome hybridization confirmed the lack of pathogenic mutations and large-scale deletions in TBX3 (12q24.21), the only gene known to be associated with UMS cases to date. Based on the results of whole-exome sequencing, 14 potential candidate variants were identified. Among them, a novel missense variant in SYNM gene (exon 1, c.173C > T, p.A58V), encoding intermediate filament protein synemin was characterized. Until the present, no association between SYNM mutations and congenital clinical syndromes has been reported. At the same time, taking into account synemin tissue-specific expression profiles and available data on abnormal knock-out mice phenotypes, we propose SYNM as a candidate gene contributing to the UMS-like phenotype. Further comprehensive functional studies are required to evaluate possible involvement of SYNM in genesis of complex heart-limb pathology.
Collapse
Affiliation(s)
- Anna Zlotina
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Artem Kiselev
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | | | - Elena Parmon
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia
- Department of Women’s and Children’s Health, Center for Molecular Medicine, Karolinska Institute, Solna, Sweden
| |
Collapse
|
12
|
Criswell S, O’Brien T, Skalli O. Presence of intermediate filament protein synemin in select sarcomas. J Histotechnol 2018. [DOI: 10.1080/01478885.2018.1438757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Sheila Criswell
- Department of Clinical Laboratory Science, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Thomas O’Brien
- Memphis Pathology Group, Department of Pathology, Methodist University Hospital, Memphis, TN, USA
| | - Omar Skalli
- Department of Biological Sciences, University of Memphis, Memphis, TN, USA
| |
Collapse
|
13
|
Abstract
Cardiac and skeletal striated muscles are intricately designed machines responsible for muscle contraction. Coordination of the basic contractile unit, the sarcomere, and the complex cytoskeletal networks are critical for contractile activity. The sarcomere is comprised of precisely organized individual filament systems that include thin (actin), thick (myosin), titin, and nebulin. Connecting the sarcomere to other organelles (e.g., mitochondria and nucleus) and serving as the scaffold to maintain cellular integrity are the intermediate filaments. The costamere, on the other hand, tethers the sarcomere to the cell membrane. Unique structures like the intercalated disc in cardiac muscle and the myotendinous junction in skeletal muscle help synchronize and transmit force. Intense investigation has been done on many of the proteins that make up these cytoskeletal assemblies. Yet the details of their function and how they interconnect have just started to be elucidated. A vast number of human myopathies are contributed to mutations in muscle proteins; thus understanding their basic function provides a mechanistic understanding of muscle disorders. In this review, we highlight the components of striated muscle with respect to their interactions, signaling pathways, functions, and connections to disease. © 2017 American Physiological Society. Compr Physiol 7:891-944, 2017.
Collapse
Affiliation(s)
- Christine A Henderson
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Christopher G Gomez
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Stefanie M Novak
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Lei Mi-Mi
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
14
|
|
15
|
García-Pelagio KP, Muriel J, O'Neill A, Desmond PF, Lovering RM, Lund L, Bond M, Bloch RJ. Myopathic changes in murine skeletal muscle lacking synemin. Am J Physiol Cell Physiol 2015; 308:C448-62. [PMID: 25567810 DOI: 10.1152/ajpcell.00331.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Diseases of striated muscle linked to intermediate filament (IF) proteins are associated with defects in the organization of the contractile apparatus and its links to costameres, which connect the sarcomeres to the cell membrane. Here we study the role in skeletal muscle of synemin, a type IV IF protein, by examining mice null for synemin (synm-null). Synm-null mice have a mild skeletal muscle phenotype. Tibialis anterior (TA) muscles show a significant decrease in mean fiber diameter, a decrease in twitch and tetanic force, and an increase in susceptibility to injury caused by lengthening contractions. Organization of proteins associated with the contractile apparatus and costameres is not significantly altered in the synm-null. Elastimetry of the sarcolemma and associated contractile apparatus in extensor digitorum longus myofibers reveals a reduction in tension consistent with an increase in sarcolemmal deformability. Although fatigue after repeated isometric contractions is more marked in TA muscles of synm-null mice, the ability of the mice to run uphill on a treadmill is similar to controls. Our results suggest that synemin contributes to linkage between costameres and the contractile apparatus and that the absence of synemin results in decreased fiber size and increased sarcolemmal deformability and susceptibility to injury. Thus synemin plays a moderate but distinct role in fast twitch skeletal muscle.
Collapse
Affiliation(s)
- Karla P García-Pelagio
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Joaquin Muriel
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Andrea O'Neill
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Patrick F Desmond
- Program in Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Richard M Lovering
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Linda Lund
- Merrick School of Business, University of Baltimore, Baltimore, Maryland; and
| | - Meredith Bond
- College of Sciences and Health Professions, Cleveland State University, Cleveland, Ohio
| | - Robert J Bloch
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland;
| |
Collapse
|
16
|
α-Synemin localizes to the M-band of the sarcomere through interaction with the M10 region of titin. FEBS Lett 2014; 588:4625-30. [PMID: 25447537 DOI: 10.1016/j.febslet.2014.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 11/03/2014] [Accepted: 11/04/2014] [Indexed: 01/26/2023]
Abstract
α-Synemin contains a unique 312 amino acid insert near the end of its C-terminal tail. Therefore we set out to determine if the insert is a site of protein-protein interaction that regulates the sub-cellular localization of this large isoform of synemin. Yeast-two hybrid analysis indicated that this region is a binding site for the M10 region of titin. This was confirmed with GST pull-down assays. Co-immunoprecipitation of endogenous proteins indicated close association of the two proteins in vivo and immunostaining of cardiomyocytes demonstrated co-localization of the proteins at the M-band of the sarcomere.
Collapse
|
17
|
Intermediate filaments and the regulation of focal adhesion. Curr Opin Cell Biol 2014; 32:13-20. [PMID: 25460777 DOI: 10.1016/j.ceb.2014.09.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 12/19/2022]
Abstract
Focal adhesions are localized actin filament-anchoring signalling centres at the cell-extracellular matrix interface. The currently emerging view is that they fulfil an all-embracing coordinating function for the entire cytoskeleton. This review highlights the tight relationship between focal adhesions and the intermediate filament cytoskeleton. We summarize the accumulating evidence for direct binding of intermediate filaments to focal adhesion components and their mutual cross-talk through signalling molecules. Examples are presented to emphasize the high degree of complexity of these interactions equipping cells with a precisely controlled machinery for context-dependent adjustment of their biomechanical properties.
Collapse
|
18
|
Li Z, Parlakian A, Coletti D, Alonso-Martin S, Hourdé C, Joanne P, Gao-Li J, Blanc J, Ferry A, Paulin D, Xue Z, Agbulut O. Synemin acts as a regulator of signalling molecules during skeletal muscle hypertrophy. J Cell Sci 2014; 127:4589-601. [PMID: 25179606 DOI: 10.1242/jcs.143164] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Synemin, a type IV intermediate filament (IF) protein, forms a bridge between IFs and cellular membranes. As an A-kinase-anchoring protein, it also provides temporal and spatial targeting of protein kinase A (PKA). However, little is known about its functional roles in either process. To better understand its functions in muscle tissue, we generated synemin-deficient (Synm(-) (/-)) mice. Synm(-) (/-) mice displayed normal development and fertility but showed a mild degeneration and regeneration phenotype in myofibres and defects in sarcolemma membranes. Following mechanical overload, Synm(-) (/-) mice muscles showed a higher hypertrophic capacity with increased maximal force and fatigue resistance compared with control mice. At the molecular level, increased remodelling capacity was accompanied by decreased myostatin (also known as GDF8) and atrogin (also known as FBXO32) expression, and increased follistatin expression. Furthermore, the activity of muscle-mass control molecules (the PKA RIIα subunit, p70S6K and CREB1) was increased in mutant mice. Finally, analysis of muscle satellite cell behaviour suggested that the absence of synemin could affect the balance between self-renewal and differentiation of these cells. Taken together, our results show that synemin is necessary to maintain membrane integrity and regulates signalling molecules during muscle hypertrophy.
Collapse
Affiliation(s)
- Zhenlin Li
- Sorbonne Universités, UPMC Univ Paris 06, UMR CNRS 8256/INSERM ERL U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Paris, F-75005 France
| | - Ara Parlakian
- Sorbonne Universités, UPMC Univ Paris 06, UMR CNRS 8256/INSERM ERL U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Paris, F-75005 France
| | - Dario Coletti
- Sorbonne Universités, UPMC Univ Paris 06, UMR CNRS 8256/INSERM ERL U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Paris, F-75005 France
| | - Sonia Alonso-Martin
- Sorbonne Universités, UPMC Univ-Paris 06, INSERM U974, CNRS UMR7215, Institut de Myologie, Paris-France
| | - Christophe Hourdé
- Sorbonne Universités, UPMC Univ Paris 06, UMR CNRS 8256/INSERM ERL U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Paris, F-75005 France
| | - Pierre Joanne
- Sorbonne Universités, UPMC Univ Paris 06, UMR CNRS 8256/INSERM ERL U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Paris, F-75005 France
| | - Jacqueline Gao-Li
- Sorbonne Universités, UPMC Univ Paris 06, UMR CNRS 8256/INSERM ERL U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Paris, F-75005 France
| | - Jocelyne Blanc
- Sorbonne Universités, UPMC Univ Paris 06, UMR CNRS 8256/INSERM ERL U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Paris, F-75005 France
| | - Arnaud Ferry
- Sorbonne Universités, UPMC Univ-Paris 06, INSERM U974, CNRS UMR7215, Institut de Myologie, Paris-France
| | - Denise Paulin
- Sorbonne Universités, UPMC Univ Paris 06, UMR CNRS 8256/INSERM ERL U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Paris, F-75005 France
| | - Zhigang Xue
- Sorbonne Universités, UPMC Univ Paris 06, UMR CNRS 8256/INSERM ERL U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Paris, F-75005 France
| | - Onnik Agbulut
- Sorbonne Universités, UPMC Univ Paris 06, UMR CNRS 8256/INSERM ERL U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Paris, F-75005 France
| |
Collapse
|
19
|
Banks GB, Combs AC, Odom GL, Bloch RJ, Chamberlain JS. Muscle structure influences utrophin expression in mdx mice. PLoS Genet 2014; 10:e1004431. [PMID: 24922526 PMCID: PMC4055409 DOI: 10.1371/journal.pgen.1004431] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/24/2014] [Indexed: 02/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscle wasting disorder caused by mutations in the dystrophin gene. To examine the influence of muscle structure on the pathogenesis of DMD we generated mdx4cv:desmin double knockout (dko) mice. The dko male mice died of apparent cardiorespiratory failure at a median age of 76 days compared to 609 days for the desmin−/− mice. An ∼2.5 fold increase in utrophin expression in the dko skeletal muscles prevented necrosis in ∼91% of 1a, 2a and 2d/x fiber-types. In contrast, utrophin expression was reduced in the extrasynaptic sarcolemma of the dko fast 2b fibers leading to increased membrane fragility and dystrophic pathology. Despite lacking extrasynaptic utrophin, the dko fast 2b fibers were less dystrophic than the mdx4cv fast 2b fibers suggesting utrophin-independent mechanisms were also contributing to the reduced dystrophic pathology. We found no overt change in the regenerative capacity of muscle stem cells when comparing the wild-type, desmin−/−, mdx4cv and dko gastrocnemius muscles injured with notexin. Utrophin could form costameric striations with α-sarcomeric actin in the dko to maintain the integrity of the membrane, but the lack of restoration of the NODS (nNOS, α-dystrobrevin 1 and 2, α1-syntrophin) complex and desmin coincided with profound changes to the sarcomere alignment in the diaphragm, deposition of collagen between the myofibers, and impaired diaphragm function. We conclude that the dko mice may provide new insights into the structural mechanisms that influence endogenous utrophin expression that are pertinent for developing a therapy for DMD. Duchenne muscular dystrophy (DMD) is a severe muscle wasting disorder caused by mutations in the dystrophin gene. Utrophin is structurally similar to dystrophin and improving its expression can prevent skeletal muscle necrosis in the mdx mouse model of DMD. Consequently, improving utrophin expression is a primary therapeutic target for treating DMD. While the downstream mechanisms that influence utrophin expression and stability are well described, the upstream mechanisms are less clear. Here, we found that perturbing the highly ordered structure of striated muscle by genetically deleting desmin from mdx mice increased utrophin expression to levels that prevented skeletal muscle necrosis. Thus, the mdx:desmin double knockout mice may prove valuable in determining the upstream mechanisms that influence utrophin expression to develop a therapy for DMD.
Collapse
Affiliation(s)
- Glen B. Banks
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - Ariana C. Combs
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
| | - Guy L. Odom
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
| | - Robert J. Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jeffrey S. Chamberlain
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
20
|
Rodler D, Sinowatz F. Expression of intermediate filaments in the Balbiani body and ovarian follicular wall of the Japanese quail (Coturnix japonica). Cells Tissues Organs 2013; 197:298-311. [PMID: 23391820 DOI: 10.1159/000346048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2012] [Indexed: 11/19/2022] Open
Abstract
In the present study, we examined the distribution of 6 groups of intermediate filaments (IFs; cytokeratins, CKs, vimentin, synemin, desmin, glial fibrillary acidic protein and lamins) in oocytes and follicular walls of the Japanese quail (Coturnix japonica) during their development using immunohistochemical and ultrastructural techniques. A distinctly vimentin- and synemin-positive Balbiani body, which is a transient accumulation of organelles (mitochondria, Golgi complex and endoplasmic reticulum) that occurs in the oocytes of all vertebrates including birds, could be detected in the oocytes of primordial and early pre-vitellogenic follicles. In larger pre-vitellogenic follicles, the Balbiani body has dispersed and the positivity of the granulosa cells appeared to concentrate in the basal portion of their cytoplasm. Our ultrastructural data demonstrated that the matrix of the Bal-biani body consists of fine IFs, which may play a role in the formation and dispersion of the Balbiani body. Of the CKs studied (panCK, CK5, CK7, CK8, CK14, CK15, CK18 and CK19), only CK5 showed a slight positive staining in both the theca externa and the Balbiani bodies of pre-vitellogenic oocytes. In conclusion, our data, which describe the changes in avian IF protein expression during folliculogenesis, suggest that the functions of the IFs (vimentin and synemin) of oocytes and follicular walls are not primarily mechanical but may be involved in the transient tethering of mitochondria in the area of the Balbiani body and in the gain of endocrine competence during the differentiation of granulosa cells.
Collapse
Affiliation(s)
- Daniela Rodler
- Department of Veterinary Sciences, Institute of Anatomy, Histology and Embryology, University of Munich, DE–80539 Munich, Germany
| | | |
Collapse
|
21
|
Desminopathies: pathology and mechanisms. Acta Neuropathol 2013; 125:47-75. [PMID: 23143191 PMCID: PMC3535371 DOI: 10.1007/s00401-012-1057-6] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Revised: 10/15/2012] [Accepted: 10/18/2012] [Indexed: 12/22/2022]
Abstract
The intermediate filament protein desmin is an essential component of the extra-sarcomeric cytoskeleton in muscle cells. This three-dimensional filamentous framework exerts central roles in the structural and functional alignment and anchorage of myofibrils, the positioning of cell organelles and signaling events. Mutations of the human desmin gene on chromosome 2q35 cause autosomal dominant, autosomal recessive, and sporadic myopathies and/or cardiomyopathies with marked phenotypic variability. The disease onset ranges from childhood to late adulthood. The clinical course is progressive and no specific treatment is currently available for this severely disabling disease. The muscle pathology is characterized by desmin-positive protein aggregates and degenerative changes of the myofibrillar apparatus. The molecular pathophysiology of desminopathies is a complex, multilevel issue. In addition to direct effects on the formation and maintenance of the extra-sarcomeric intermediate filament network, mutant desmin affects essential protein interactions, cell signaling cascades, mitochondrial functions, and protein quality control mechanisms. This review summarizes the currently available data on the epidemiology, clinical phenotypes, myopathology, and genetics of desminopathies. In addition, this work provides an overview on the expression, filament formation processes, biomechanical properties, post-translational modifications, interaction partners, subcellular localization, and functions of wild-type and mutant desmin as well as desmin-related cell and animal models.
Collapse
|
22
|
Pitre A, Davis N, Paul M, Orr AW, Skalli O. Synemin promotes AKT-dependent glioblastoma cell proliferation by antagonizing PP2A. Mol Biol Cell 2012; 23:1243-53. [PMID: 22337773 PMCID: PMC3315805 DOI: 10.1091/mbc.e11-08-0685] [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] [Indexed: 12/22/2022] Open
Abstract
Synemin is an intermediate filament protein present in glioblastomas (GBMs) but not in normal brain. In GBM cells synemin interacts with and antagonizes PP2A, which is the phosphatase dephosphorylating Akt. This maintains the phosphorylation status of Akt sites that are substrates for PDPK1 and mTORc2, thereby fostering proliferation. The intermediate filament protein synemin is present in astrocyte progenitors and glioblastoma cells but not in mature astrocytes. Here we demonstrate a role for synemin in enhancing glioblastoma cell proliferation and clonogenic survival, as synemin RNA interference decreased both behaviors by inducing G1 arrest along with Rb hypophosphorylation and increased protein levels of the G1/S inhibitors p21Cip1 and p27Kip1. Akt involvement was demonstrated by decreased phosphorylation of its substrate, p21Cip1, and reduced Akt catalytic activity and phosphorylation at essential activation sites. Synemin silencing, however, did not affect the activities of PDPK1 and mTOR complex 2, which directly phosphorylate Akt activation sites, but instead enhanced the activity of the major regulator of Akt dephosphorylation, protein phosphatase type 2A (PP2A). This was accompanied by changes in PP2A subcellular distribution resulting in increased physical interactions between PP2A and Akt, as shown by proximity ligation assays (PLAs). PLAs and immunoprecipitation experiments further revealed that synemin and PP2A form a protein complex. In addition, treatment of synemin-silenced cells with the PP2A inhibitor cantharidic acid resulted in proliferation and pAkt and pRb levels similar to those of controls. Collectively these results indicate that synemin positively regulates glioblastoma cell proliferation by helping sequester PP2A away from Akt, thereby favoring Akt activation.
Collapse
Affiliation(s)
- Aaron Pitre
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | | | | | | | | |
Collapse
|
23
|
Dynamic expression of synemin isoforms in mouse embryonic stem cells and neural derivatives. BMC Cell Biol 2011; 12:51. [PMID: 22107957 PMCID: PMC3233510 DOI: 10.1186/1471-2121-12-51] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 11/23/2011] [Indexed: 01/04/2023] Open
Abstract
Background Intermediate filaments (IFs) are major components of the mammalian cytoskeleton and expressed in cell-type-specific patterns. Morphological changes during cell differentiation are linked to IF network remodeling. However, little is known concerning the presence and the role of IFs in embryonic stem (ES) cells and during their differentiation. Results We have examined the expression profile of synemin isoforms in mouse pluripotent ES cells and during their neural differentiation induced by retinoic acid. Using RT-PCR, Western blotting and immunostaining, we show that synemin M is present at both mRNA and protein levels in undifferentiated ES cells as early as pluripotency factor Oct-3/4 and IF keratin 8. Synemin H was produced only in neural precursors when neural differentiation started, concurrently with synemin M, nestin and glial fibrillary acidic protein. However, both synemin H and M were restricted to the progenitor line during the neural differentiation program. Our in vivo analysis also confirmed the expression of synemins H/M in multipotent neural stem cells in the subventricular zone of the adult brain, a neurogenic germinal niche of the mice. Knocking down synemin in ES cells by shRNA lentiviral particles transduction has no influence on expression of Oct4, Nanog and SOX2, but decreased keratin 8 expression. Conclusions Our study shows a developmental stage specific regulation of synemin isoforms in ES cells and its neural derivatives. These findings represent the first evidence that synemins could potentially be useful markers for distinguishing multipotent ES cells from undifferentiated neural stem cells and more committed progenitor cells.
Collapse
|
24
|
Lund LM, Kerr JP, Lupinetti J, Zhang Y, Russell MA, Bloch RJ, Bond M. Synemin isoforms differentially organize cell junctions and desmin filaments in neonatal cardiomyocytes. FASEB J 2011; 26:137-48. [PMID: 21982947 DOI: 10.1096/fj.10-179408] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Intermediate filaments (IFs) in cardiomyocytes consist primarily of desmin, surround myofibrils at Z disks, and transmit forces from the contracting myofilaments to the cell surface through costameres at the sarcolemma and desmosomes at intercalated disks. Synemin is a type IV IF protein that forms filaments with desmin and also binds α-actinin and vinculin. Here we examine the roles and expression of the α and β forms of synemin in developing rat cardiomyocytes. Quantitative PCR showed low levels of expression for both synemin mRNAs, which peaked at postnatal day 7. Synemin was concentrated at sites of cell-cell adhesion and at Z disks in neonatal cardiomyocytes. Overexpression of the individual isoforms showed that α-synemin preferentially localized to cell-cell junctions, whereas β-synemin was primarily at the level of Z disks. An siRNA targeted to both synemin isoforms reduced protein expression in cardiomyocytes by 70% and resulted in a failure of desmin to align with Z disks and disrupted cell-cell junctions, with no effect on sarcomeric organization. Solubility assays showed that β-synemin was soluble and interacted with sarcomeric α-actinin by coimmunoprecipitation, while α-synemin and desmin were insoluble. We conclude that β-synemin mediates the association of desmin IFs with Z disks, whereas α-synemin stabilizes junctional complexes between cardiomyocytes.
Collapse
Affiliation(s)
- Linda M Lund
- Department of Physiology, University of Maryland, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | |
Collapse
|
25
|
Dupin I, Etienne-Manneville S. Nuclear positioning: mechanisms and functions. Int J Biochem Cell Biol 2011; 43:1698-707. [PMID: 21959251 DOI: 10.1016/j.biocel.2011.09.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 09/10/2011] [Accepted: 09/15/2011] [Indexed: 10/17/2022]
Abstract
The nucleus is the largest organelle in the cell and its position is dynamically controlled in space and time, although the functional significance of this dynamic regulation is not always clear. Nuclear movements are mediated by the cytoskeleton which transmits pushing or pulling forces onto the nuclear envelope. Recent studies have shed light on the mechanisms regulating nuclear positioning inside the cell. While microtubules have been known for a long time to be key players in nuclear positioning, the actin and cytoplasmic intermediate filament cytoskeletons have been implicated in this function more recently and various molecular links between the nuclear envelope and cytoplasmic elements have been identified. In this review, we summarize the recent advances in our understanding of the molecular mechanisms involved in the regulation of nuclear localization in various animal cells and give an overview of the evidence suggesting a crucial role of nuclear positioning in cell polarity and physiology and the consequences of nuclear mispositioning in human pathologies.
Collapse
Affiliation(s)
- Isabelle Dupin
- Institut Pasteur, Cell Polarity, Migration and Cancer Unit and CNRS URA 2582, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | | |
Collapse
|
26
|
Seto JT, Lek M, Quinlan KGR, Houweling PJ, Zheng XF, Garton F, MacArthur DG, Raftery JM, Garvey SM, Hauser MA, Yang N, Head SI, North KN. Deficiency of α-actinin-3 is associated with increased susceptibility to contraction-induced damage and skeletal muscle remodeling. Hum Mol Genet 2011; 20:2914-27. [PMID: 21536590 DOI: 10.1093/hmg/ddr196] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Sarcomeric α-actinins (α-actinin-2 and -3) are a major component of the Z-disk in skeletal muscle, where they crosslink actin and other structural proteins to maintain an ordered myofibrillar array. Homozygosity for the common null polymorphism (R577X) in ACTN3 results in the absence of fast fiber-specific α-actinin-3 in ∼20% of the general population. α-Actinin-3 deficiency is associated with decreased force generation and is detrimental to sprint and power performance in elite athletes, suggesting that α-actinin-3 is necessary for optimal forceful repetitive muscle contractions. Since Z-disks are the structures most vulnerable to eccentric damage, we sought to examine the effects of α-actinin-3 deficiency on sarcomeric integrity. Actn3 knockout mouse muscle showed significantly increased force deficits following eccentric contraction at 30% stretch, suggesting that α-actinin-3 deficiency results in an increased susceptibility to muscle damage at the extremes of muscle performance. Microarray analyses demonstrated an increase in muscle remodeling genes, which we confirmed at the protein level. The loss of α-actinin-3 and up-regulation of α-actinin-2 resulted in no significant changes to the total pool of sarcomeric α-actinins, suggesting that alterations in fast fiber Z-disk properties may be related to differences in functional protein interactions between α-actinin-2 and α-actinin-3. In support of this, we demonstrated that the Z-disk proteins, ZASP, titin and vinculin preferentially bind to α-actinin-2. Thus, the loss of α-actinin-3 changes the overall protein composition of fast fiber Z-disks and alters their elastic properties, providing a mechanistic explanation for the loss of force generation and increased susceptibility to eccentric damage in α-actinin-3-deficient individuals.
Collapse
Affiliation(s)
- Jane T Seto
- Institute for Neuroscience and Muscle Research, The Children's Hospital at Westmead, Locked Bag 4001, Sydney, NSW 2145, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Liu YH, Cheng CC, Lai YS, Chao WT, Pei RJ, Hsu YH, Ho CC. Synemin down-regulation in human hepatocellular carcinoma does not destabilize cytoskeletons in vivo. Biochem Biophys Res Commun 2010; 404:488-93. [PMID: 21144834 DOI: 10.1016/j.bbrc.2010.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Accepted: 12/02/2010] [Indexed: 11/17/2022]
Abstract
Synemin is a large intermediate filament protein that has been identified in all types of muscle cells. It plays a role in human muscle diseases; however, the role of synemin in tumor cell transformation has rarely been investigated. Because hepatocellular carcinoma cells are morphologically different from normal human hepatocytes, we hypothesized that altered synemin expression and cytoskeletal disorganization might underlie this pleomorphic transformation. To test this hypothesis, we studied synemin expression in hepatocellular carcinoma and liver tissues by immunohistochemistry and immunoblotting. In addition, we analyzed the expression level and organization of all cytoskeletal elements after synemin knock-down in human Chang liver cells. Previously we found that plectin knock-down in human Chang liver cells causes a reduction in cytokeratin 18 expression with effects on intermediate filament disorganization and altered cellular morphology. In this study we also compared the effects of synemin knock-down and plectin knock-down on the cytoskeleton expression and organization. The results revealed that synemin expression was down-regulated in human hepatocellular carcinoma compared with normal liver, which is similar to the plectin expression. Surprisingly, the expression of cytoskeletal elements (cytokeratin 18, actin and tubulin) was not influenced by synemin knock-down in human Chang liver cells. The organization of cytoskeletal networks was also unaltered after synemin knock-down. In conclusion, both plectin and synemin are down-regulated in human hepatocellular carcinoma in vivo and transformed human liver cell in vitro. However, the mechanism of cell transformation caused by synemin knock-down is different from that of plectin knock-down. Plectin, but not synemin, knock-down provoked liver cell transformation via suppressing cytokeratin 18 expression and disrupting intermediate filament networks. Synemin knock-down did not influence the cytoskeleton expression and organization of human Chang liver cells.
Collapse
Affiliation(s)
- Yi-Hsiang Liu
- Department of Pathology, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | | | | | | | | | | | | |
Collapse
|
28
|
Intermediate filament dynamics and breast cancer: aberrant promoter methylation of the Synemin gene is associated with early tumor relapse. Oncogene 2010; 29:4814-25. [PMID: 20543860 DOI: 10.1038/onc.2010.229] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Synemin (SYNM) is a type IV intermediate filament that has recently been shown to interact with the LIM domain protein zyxin, thereby possibly modulating cell adhesion and cell motility. Owing to this multiplicity of potential functions relevant to cancer development, we initiated a study to decipher SYNM expression and regulation in benign human breast tissue and breast cancer. Dot blot array analysis showed significant SYNM mRNA downregulation in 86% (n=100, P<0.001) of breast cancers compared with their normal tissue counterparts, a result that was confirmed by real-time PCR analysis (n=36, P<0.0001). Immunohistochemistry analysis showed abundant SYNM protein expression in healthy myoepithelial breast cells, whereas SYNM expression loss was evident in 57% (n=37, P<0.001) of breast cancer specimens. Next, we analyzed methylation of the SYNM promoter to clarify whether the SYNM gene can be silenced by epigenetic means. Indeed, methylation-specific PCR analysis showed tumor-specific SYNM promoter methylation in 27% (n=195) of breast cancers. As expected, SYNM promoter methylation was tightly associated (P<0.0001) with SYNM expression loss. In-depth analysis of the SYNM promoter by pyrosequencing showed extensive CpG methylation of DNA elements supposed to regulate gene transcription. Demethylating treatment of SYNM methylated breast cancer cell lines with 5-aza-2-deoxycytidine clearly reestablished the SYNM expression. Statistical analysis of the patient cohort showed a close association between SYNM promoter methylation and unfavorable recurrence-free survival (hazard ratio=2.941, P=0.0282). Furthermore, SYNM methylation positively correlated with lymph node metastases (P=0.0177) and advanced tumor grade (P=0.0275), suggesting that SYNM methylation is associated with aggressive forms of breast cancer. This is the first study on the epigenetic regulation of the SYNM gene in a cancer entity. We provide first hints that SYNM could represent a novel putative breast tumor suppressor gene that is prone to epigenetic silencing. SYNM promoter methylation may become a useful predictive biomarker to stratify breast cancer patients' risk for tumor relapse.
Collapse
|
29
|
Steiner-Champliaud MF, Schneider Y, Favre B, Paulhe F, Praetzel-Wunder S, Faulkner G, Konieczny P, Raith M, Wiche G, Adebola A, Liem RK, Langbein L, Sonnenberg A, Fontao L, Borradori L. BPAG1 isoform-b: Complex distribution pattern in striated and heart muscle and association with plectin and α-actinin. Exp Cell Res 2010; 316:297-313. [DOI: 10.1016/j.yexcr.2009.11.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Revised: 11/13/2009] [Accepted: 11/16/2009] [Indexed: 10/20/2022]
|
30
|
Izmiryan A, Peltekian E, Paulin D, Li ZL, Xue ZG. Synemin Isoforms in Astroglial and Neuronal Cells from Human Central Nervous System. Neurochem Res 2009; 35:881-7. [DOI: 10.1007/s11064-009-0111-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2009] [Indexed: 10/20/2022]
|
31
|
Sun N, Huiatt TW, Paulin D, Li Z, Robson RM. Synemin interacts with the LIM domain protein zyxin and is essential for cell adhesion and migration. Exp Cell Res 2009; 316:491-505. [PMID: 19853601 DOI: 10.1016/j.yexcr.2009.10.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 10/14/2009] [Accepted: 10/16/2009] [Indexed: 01/12/2023]
Abstract
Synemin is a unique cytoplasmic intermediate filament protein for which there is limited understanding of its exact cellular functions. The single human synemin gene encodes at least two splice variants named alpha-synemin and beta-synemin, with the larger alpha-synemin containing an additional 312 amino acid insert within the C-terminal tail domain. We report herein that, by using the entire tail domain of the smaller beta-synemin as the bait in a yeast two-hybrid screen of a human skeletal muscle cDNA library, the LIM domain protein zyxin was identified as an interaction partner for human synemin. The synemin binding site in human zyxin was subsequently mapped to the C-terminal three tandem LIM-domain repeats, whereas the binding site for zyxin within beta-synemin is within the C-terminal 332 amino acid region (SNbetaTII) at the end of the long tail domain. Transient expression of SNbetaTII within mammalian cells markedly reduced zyxin protein level, blocked localization of zyxin at focal adhesion sites and resulted in decreased cell adhesion and increased motility. Knockdown of synemin expression with siRNAs within mammalian cells resulted in significantly compromised cell adhesion and cell motility. Our results suggest that synemin participates in focal adhesion dynamics and is essential for cell adhesion and migration.
Collapse
Affiliation(s)
- Ning Sun
- Muscle Biology Group, Department of Biochemistry, Biophysics and Molecular Biology and of Animal Science, Iowa State University, Ames, 3110 Molecular Biology Bldg, IA 50011-3260, USA
| | | | | | | | | |
Collapse
|
32
|
Mauban JRH, O'Donnell M, Warrier S, Manni S, Bond M. AKAP-scaffolding proteins and regulation of cardiac physiology. Physiology (Bethesda) 2009; 24:78-87. [PMID: 19364910 DOI: 10.1152/physiol.00041.2008] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
A kinase anchoring proteins (AKAPs) compose a growing list of diverse but functionally related proteins defined by their ability to bind to the regulatory subunit of protein kinase A. AKAPs perform an integral role in the spatiotemporal modulation of a multitude of cellular signaling pathways. This review highlights the extensive role of AKAPs in cardiac excitation/contraction coupling and cardiac physiology. The literature shows that particular AKAPs are involved in cardiac Ca(2+) influx, release, reuptake, and myocyte repolarization. Studies have also suggested roles for AKAPs in cardiac remodeling. Transgenic studies show functional effects of AKAPs, not only in the cardiovascular system but in other organ systems as well.
Collapse
Affiliation(s)
- J R H Mauban
- Departments of Physiology, University of Maryland Baltimore, Baltimore, Maryland, USA
| | | | | | | | | |
Collapse
|
33
|
Izmiryan A, Franco CA, Paulin D, Li Z, Xue Z. Synemin isoforms during mouse development: Multiplicity of partners in vascular and neuronal systems. Exp Cell Res 2009; 315:769-83. [DOI: 10.1016/j.yexcr.2008.12.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 11/21/2008] [Accepted: 12/07/2008] [Indexed: 11/25/2022]
|
34
|
Khanamiryan L, Li Z, Paulin D, Xue Z. Self-Assembly Incompetence of Synemin Is Related to the Property of Its Head and Rod Domains. Biochemistry 2008; 47:9531-9. [DOI: 10.1021/bi800912w] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Luiza Khanamiryan
- UPMC Univ Paris 6, UMR 7079, Paris, France, and CNRS UMR 7079, Paris, France
| | - Zhenlin Li
- UPMC Univ Paris 6, UMR 7079, Paris, France, and CNRS UMR 7079, Paris, France
| | - Denise Paulin
- UPMC Univ Paris 6, UMR 7079, Paris, France, and CNRS UMR 7079, Paris, France
| | - Zhigang Xue
- UPMC Univ Paris 6, UMR 7079, Paris, France, and CNRS UMR 7079, Paris, France
| |
Collapse
|
35
|
McCullagh KJA, Edwards B, Kemp MW, Giles LC, Burgess M, Davies KE. Analysis of skeletal muscle function in the C57BL6/SV129 syncoilin knockout mouse. Mamm Genome 2008; 19:339-51. [PMID: 18594912 PMCID: PMC2515546 DOI: 10.1007/s00335-008-9120-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Accepted: 05/28/2008] [Indexed: 11/28/2022]
Abstract
Syncoilin is a 64-kDa intermediate filament protein expressed in skeletal muscle and enriched at the perinucleus, sarcolemma, and myotendinous and neuromuscular junctions. Due to its pattern of cellular localization and binding partners, syncoilin is an ideal candidate to be both an important structural component of myocytes and a potential mediator of inherited myopathies. Here we present a report of a knockout mouse model for syncoilin and the results of an investigation into the effect of a syncoilin null state on striated muscle function in 6–8-week-old mice. An analysis of proteins known to associate with syncoilin showed that ablation of syncoilin had no effect on absolute expression or spatial localization of desmin or alpha dystrobrevin. Our syncoilin-null animal exhibited no differences in cardiotoxin-induced muscle regeneration, voluntary wheel running, or enforced treadmill exercise capacity, relative to wild-type controls. Finally, a mechanical investigation of isolated soleus and extensor digitorum longus indicated a potential differential reduction in muscle strength and resilience. We are the first to present data identifying an increased susceptibility to muscle damage in response to an extended forced exercise regime in syncoilin-deficient muscle. This study establishes a second viable syncoilin knockout model and highlights the importance of further investigations to determine the role of syncoilin in skeletal muscle.
Collapse
Affiliation(s)
- Karl J A McCullagh
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | | | | | | | | | | |
Collapse
|
36
|
Hijikata T, Nakamura A, Isokawa K, Imamura M, Yuasa K, Ishikawa R, Kohama K, Takeda S, Yorifuji H. Plectin 1 links intermediate filaments to costameric sarcolemma through β-synemin, α-dystrobrevin and actin. J Cell Sci 2008; 121:2062-74. [DOI: 10.1242/jcs.021634] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In skeletal muscles, the sarcolemma is possibly stabilized and protected against contraction-imposed stress by intermediate filaments (IFs) tethered to costameric sarcolemma. Although there is emerging evidence that plectin links IFs to costameres through dystrophin-glycoprotein complexes (DGC), the molecular organization from plectin to costameres still remains unclear. Here, we show that plectin 1, a plectin isoform expressed in skeletal muscle, can interact with β-synemin, actin and a DGC component, α-dystrobrevin, in vitro. Ultrastructurally, β-synemin molecules appear to be incorporated into costameric dense plaques, where they seem to serve as actin-associated proteins rather than IF proteins. In fact, they can bind actin and α-dystrobrevin in vitro. Moreover, in vivo immunoprecipitation analyses demonstrated that β-synemin- and plectin-immune complexes from lysates of muscle light microsomes contained α-dystrobrevin, dystrophin, nonmuscle actin, metavinculin, plectin and β-synemin. These findings suggest a model in which plectin 1 interacts with DGC and integrin complexes directly, or indirectly through nonmuscle actin and β-synemin within costameres. The DGC and integrin complexes would cooperate to stabilize and fortify the sarcolemma by linking the basement membrane to IFs through plectin 1, β-synemin and actin. Besides, the two complexes, together with plectin and IFs, might have their own functions as platforms for distinct signal transduction.
Collapse
Affiliation(s)
- Takao Hijikata
- Department of Anatomy and Cell Biology, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo 202-8585, Japan
| | - Akio Nakamura
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Keitaro Isokawa
- Department of Anatomy, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| | - Michihiro Imamura
- Department of Molecular Therapy, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
| | - Katsutoshi Yuasa
- Department of Anatomy and Cell Biology, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo 202-8585, Japan
| | - Ryoki Ishikawa
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Kazuhiro Kohama
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Shinichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
| | - Hiroshi Yorifuji
- Department of Anatomy, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| |
Collapse
|
37
|
Pan Y, Jing R, Pitre A, Williams BJ, Skalli O. Intermediate filament protein synemin contributes to the migratory properties of astrocytoma cells by influencing the dynamics of the actin cytoskeleton. FASEB J 2008; 22:3196-206. [PMID: 18509200 DOI: 10.1096/fj.08-106187] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have shown previously that, in astrocytoma cells, synemin is present at the leading edge, an unusual localization for an intermediate filament (IF) protein. Here, we report that synemin down-regulation with specific small hairpin RNAs (shRNAs) sharply decreased the migration of astrocytoma cells. The presence of synemin at the leading edge also correlated with a high migratory potential, as shown by comparing astrocytoma cells to carcinoma cells without synemin at the leading edge. Synemin-silenced astrocytoma cells were smaller and spread more slowly than controls. In addition, synemin silencing reduced proliferation without increasing apoptosis. The adhesion to substratum and distribution of vinculin in focal contacts of synemin-silenced astrocytoma cells were similar to those of controls. Synemin-silenced cells, however, exhibited a reduction in the amount of filamentous (F) -actin and of alpha-actinin, but not of vinculin, associated with F-actin. Altogether, these results demonstrate that synemin is important for the malignant behavior of astrocytoma cells and that it contributes to the high motility of these cells by modulating the dynamics of alpha-actinin and actin.
Collapse
Affiliation(s)
- Yihang Pan
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, 1501 Kings Hwy., Shreveport, LA 71130, USA
| | | | | | | | | |
Collapse
|
38
|
Sun N, Critchley DR, Paulin D, Li Z, Robson RM. Identification of a repeated domain within mammalian alpha-synemin that interacts directly with talin. Exp Cell Res 2008; 314:1839-49. [PMID: 18342854 DOI: 10.1016/j.yexcr.2008.01.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Revised: 01/25/2008] [Accepted: 01/31/2008] [Indexed: 12/21/2022]
Abstract
The type VI intermediate filament (IF) protein synemin is a unique member of the IF protein superfamily. Synemin associates with the major type III IF protein desmin forming heteropolymeric intermediate filaments (IFs) within developed mammalian striated muscle cells. These IFs encircle and link all adjacent myofibrils together at their Z-lines, as well as link the Z-lines of the peripheral layer of cellular myofibrils to the costameres located periodically along and subjacent to the sarcolemma. Costameres are multi-protein assemblies enriched in the cytoskeletal proteins vinculin, alpha-actinin, and talin. We report herein a direct interaction of human alpha-synemin with the cytoskeletal protein talin by protein-protein interaction assays. The 312 amino acid insert (SNTIII) present only within alpha-synemin binds to the rod domain of talin in vitro and co-localizes with talin at focal adhesion sites within mammalian muscle cells. Confocal microscopy studies showed that synemin co-localizes with talin within the costameres of human skeletal muscle cells. Analysis of the primary sequences of human alpha- and beta-synemins revealed that SNTIII is composed of seven tandem repeats, each containing a specific Ser/Thr-X-Arg-His/Gln (S/T-X-R-H/Q) motif. Our results suggest human alpha-synemin plays an essential role in linking the heteropolymeric IFs to adherens-type junctions, such as the costameres within mammalian striated muscle cells, via its interaction with talin, thereby helping provide mechanical integration for the muscle cell cytoskeleton.
Collapse
Affiliation(s)
- Ning Sun
- Muscle Biology Group, Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, IA 50011-3260, USA
| | | | | | | | | |
Collapse
|
39
|
Abstract
Anchorage of the contractile actomyosin apparatus to the plasma membrane at discrete sites in muscle and non-muscle cells enables the transmission and conversion of force into work, such as muscle contraction and membrane deformation to regulate cell and tissue shape. Assembly, stabilization and turnover of adhesion sites are complex processes that involve structural components, a variety of signalling and adapter molecules, diverse kinases and phosphatases, and phospholipids. The dynamic turnover of adhesions also requires the frequent interaction with other filament systems of the cytoskeleton, in particular with microtubules. How the delivery and activation of all the required components is co-ordinated, however, remains to be fully understood. In the current issue of Biochemical Journal, Sun et al. provide evidence that a specific exon that is exclusively present in the alpha variant of the type IV intermediate filament protein synemin interacts directly with the focal adhesion protein vinculin in its active state. Interaction of adhesion components with intermediate filaments could serve as a general mechanism to regulate cell- and tissue-specific cytoskeleton-membrane attachment.
Collapse
|
40
|
Abstract
Synemin is a very large, unique member of the IF (intermediate filament) protein superfamily. Association of synemin with the major IF proteins, desmin and/or vimentin, within muscle cells forms heteropolymeric IFs. We have previously identified interactions of avian synemin with alpha-actinin and vinculin. Avian synemin, however, is expressed as only one form, whereas human synemin is expressed as two major splice variants, namely alpha- and beta-synemins. The larger alpha-synemin contains an additional 312-amino-acid insert (termed SNTIII) located near the end of the long C-terminal tail domain. Whether alpha- and beta-synemins have different cellular functions is unclear. In the present study we show, by in vitro protein-protein interaction assays, that SNTIII interacts directly with both vinculin and metavinculin. Furthermore, SNTIII interacts with vinculin in vivo, and this association is promoted by PtdIns(4,5)P(2). SNTIII also specifically co-localizes with vinculin within focal adhesions when transiently expressed in mammalian cells. In contrast, other regions of synemin show distinct localization patterns in comparison with those of SNTIII, without labelling focal adhesions. Our results indicate that alpha-synemin, but not beta-synemin, interacts with both vinculin and metavinculin, thereby linking the heteropolymeric IFs to adhesion-type junctions, such as the costameres located within human striated muscle cells.
Collapse
|
41
|
Abstract
The intermediate filament (IF) network is one of the three cytoskeletal systems in smooth muscle. The type III IF proteins vimentin and desmin are major constituents of the network in smooth muscle cells and tissues. Lack of vimentin or desmin impairs contractile ability of various smooth muscle preparations, implying their important role for smooth muscle force development. The IF framework has long been viewed as a fixed cytostructure that solely provides mechanical integrity for the cell. However, recent studies suggest that the IF cytoskeleton is dynamic in mammalian cells in response to various external stimulation. In this review, the structure and biological properties of IF proteins in smooth muscle are summarized. The role of IF proteins in the modulation of smooth muscle force development and redistribution/translocation of signaling partners (such as p130 Crk-associated substrate, CAS) is depicted. This review also summarizes our latest understanding on how the IF network may be regulated in smooth muscle.
Collapse
Affiliation(s)
- Dale D Tang
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
42
|
Stone MR, O'Neill A, Lovering RM, Strong J, Resneck WG, Reed PW, Toivola DM, Ursitti JA, Omary MB, Bloch RJ. Absence of keratin 19 in mice causes skeletal myopathy with mitochondrial and sarcolemmal reorganization. J Cell Sci 2007; 120:3999-4008. [PMID: 17971417 DOI: 10.1242/jcs.009241] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intermediate filaments, composed of desmin and of keratins, play important roles in linking contractile elements to each other and to the sarcolemma in striated muscle. We examined the contractile properties and morphology of fast-twitch skeletal muscle from mice lacking keratin 19. Tibialis anterior muscles of keratin-19-null mice showed a small but significant decrease in mean fiber diameter and in the specific force of tetanic contraction, as well as increased plasma creatine kinase levels. Costameres at the sarcolemma of keratin-19-null muscle, visualized with antibodies against spectrin or dystrophin, were disrupted and the sarcolemma was separated from adjacent myofibrils by a large gap in which mitochondria accumulated. The costameric dystrophin-dystroglycan complex, which co-purified with gamma-actin, keratin 8 and keratin 19 from striated muscles of wild-type mice, co-purified with gamma-actin but not keratin 8 in the mutant. Our results suggest that keratin 19 in fast-twitch skeletal muscle helps organize costameres and links them to the contractile apparatus, and that the absence of keratin 19 disrupts these structures, resulting in loss of contractile force, altered distribution of mitochondria and mild myopathy. This is the first demonstration of a mammalian phenotype associated with a genetic perturbation of keratin 19.
Collapse
Affiliation(s)
- Michele R Stone
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yin H, Zhang X, Wang J, Yin W, Zhang G, Wang S, Liu Q. Downregulation of desmuslin in primary vein incompetence. J Vasc Surg 2007; 43:372-8. [PMID: 16476617 DOI: 10.1016/j.jvs.2005.10.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2005] [Accepted: 10/01/2005] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Primary vein incompetence is one of the most common diseases of the peripheral veins, but its pathogenesis is unknown. These veins present obvious congenital defects, and examination of gene expression profiles of the incompetent vein specimens may provide important clues. The aim of this study was to screen for genes affecting the primary vein incompetence phenotype and test the differential expression of certain genes. METHODS We compared gene expression profiles of valvular areas from incompetent and normal great saphenous veins at the saphenofemoral junctions by fluorescent differential display reverse-transcription polymerase chain reaction (FDD RT-PCR). Differentially expressed complimentary DNAs (cDNAs) were confirmed by Northern blotting and semi-quantitative RT-PCR. Similarity of the cDNAs sequences to GenBank sequences was determined. Gene expression status was then determined by Western blot analysis and immunohistochemical techniques. RESULTS There were >30 differentially expressed cDNA bands. Sequence analysis revealed that a cDNA fragment obviously downregulated in incompetent great saphenous vein was a portion of the messenger RNA (mRNA) encoding desmuslin, a newly discovered intermittent filament protein. Northern blotting and semi-quantitative RT-PCR analysis revealed a similar mRNA expression profile of the desmuslin gene in other samples. Western blotting and immunohistochemical techniques localized the desmuslin protein mainly in the cytoplasm of venous smooth muscle cells. The amount of desmuslin was greatly decreased in the smooth muscle cells of incompetent veins. CONCLUSIONS The expression of many genes is altered in primary vein incompetence. Up- or downregulation of these genes may be involved in the pathogenesis of this disease. Desmuslin expression is downregulated in the abnormal veins. Its effect on the integrity of smooth muscle cells might be related to malformation of the vein wall. Further studies are needed to investigate other differentially expressed cDNAs and the exact role of desmuslin in this disease. CLINICAL RELEVANCE Primary vein incompetence is a frequent and refractory disease of the peripheral veins. Exploring its pathogenesis may enhance our comprehension and management of this disease. We used reliable techniques to detect disease-related genes and confirmed downregulation of desmuslin in abnormal veins. Alteration of these genes might be used as disease markers or gene therapy targets.
Collapse
Affiliation(s)
- Henghui Yin
- Vascular Surgery Institute, Department of Vascular Surgery, The First Affiliated Hospital, Guangzhou, Peoples Republic of China
| | | | | | | | | | | | | |
Collapse
|
44
|
Guérette D, Khan PA, Savard PE, Vincent M. Molecular evolution of type VI intermediate filament proteins. BMC Evol Biol 2007; 7:164. [PMID: 17854500 PMCID: PMC2075511 DOI: 10.1186/1471-2148-7-164] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Accepted: 09/13/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tanabin, transitin and nestin are type VI intermediate filament (IF) proteins that are developmentally regulated in frogs, birds and mammals, respectively. Tanabin is expressed in the growth cones of embryonic vertebrate neurons, whereas transitin and nestin are found in myogenic and neurogenic cells. Another type VI IF protein, synemin, is expressed in undifferentiated and mature muscle cells of birds and mammals. In addition to an IF-typical alpha-helical core domain, type VI IF proteins are characterized by a long C-terminal tail often containing distinct repeated motifs. The molecular evolution of type VI IF proteins remains poorly studied. RESULTS To examine the evolutionary history of type VI IF proteins, sequence comparisons, BLAST searches, synteny studies and phylogenic analyses were performed. This study provides new evidence that tanabin, transitin and nestin are indeed orthologous type VI IF proteins. It demonstrates that tanabin, transitin and nestin genes share intron positions and sequence identities, have a similar chromosomal context and display closely related positions in phylogenic analyses. Despite this homology, fast evolution rates of their C-terminal extremity have caused the appearance of repeated motifs with distinct biological activities. In particular, our in silico and in vitro analyses of their tail domain have shown that (avian) transitin, but not (mammalian) nestin, contains a repeat domain displaying nucleotide hydrolysis activity. CONCLUSION These analyses of the evolutionary history of the IF proteins fit with a model in which type VI IFs form a branch distinct from NF proteins and are composed of two major proteins: synemin and nestin orthologs. Rapid evolution of the C-terminal extremity of nestin orthologs could be responsible for their divergent functions.
Collapse
Affiliation(s)
- Dominique Guérette
- CREFSIP and Département de médecine, Pavillon Charles-Eugène-Marchand, Université Laval, Québec, G1K 7P4, Canada
| | - Paul A Khan
- Unité de recherche en pédiatrie, Centre de recherche du CHUL, Université Laval, Québec, G1V 4G2, Canada
| | - Pierre E Savard
- Unité de recherche en Neurosciences, Centre de recherche du CHUL, Université Laval, Québec, G1V 4G2, Canada
| | - Michel Vincent
- CREFSIP and Département de médecine, Pavillon Charles-Eugène-Marchand, Université Laval, Québec, G1K 7P4, Canada
| |
Collapse
|
45
|
Hanft LM, Bogan DJ, Mayer U, Kaufman SJ, Kornegay JN, Ervasti JM. Cytoplasmic gamma-actin expression in diverse animal models of muscular dystrophy. Neuromuscul Disord 2007; 17:569-74. [PMID: 17475492 PMCID: PMC1993539 DOI: 10.1016/j.nmd.2007.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Revised: 02/26/2007] [Accepted: 03/05/2007] [Indexed: 11/23/2022]
Abstract
We recently showed that cytoplasmic gamma-actin (gamma(cyto)-actin) is dramatically elevated in striated muscle of dystrophin-deficient mdx mice. Here, we demonstrate that gamma(cyto)-actin is markedly increased in golden retriever muscular dystrophy (GRMD), which better recapitulates the dystrophinopathy phenotype in humans. Gamma(cyto)-Actin was also elevated in muscle from alpha-sarcoglycan null mice, but not in several other dystrophic animal models, including mice deficient in beta-sarcoglycan, alpha-dystrobrevin, laminin-2, or alpha7 integrin. Muscle from mice lacking dystrophin and utrophin also expressed elevated gamma(cyto)-actin, which was not restored to normal by transgenic overexpression of alpha7 integrin. However, gamma(cyto)-actin was further elevated in skeletal muscle from GRMD animals treated with the glucocorticoid prednisone at doses shown to improve the dystrophic phenotype and muscle function. These data suggest that elevated gamma(cyto)-actin is part of a compensatory cytoskeletal remodeling program that may partially stabilize dystrophic muscle in some cases where the dystrophin-glycoprotein complex is compromised.
Collapse
Affiliation(s)
- Laurin M. Hanft
- Department of Physiology, University of Wisconsin, Madison, WI
| | - Daniel J. Bogan
- College of Veterinary Medicine and Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO
| | - Ulrike Mayer
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Stephen J. Kaufman
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL
| | - Joe N. Kornegay
- College of Veterinary Medicine and Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO
| | | |
Collapse
|
46
|
Oshima RG. Intermediate filaments: a historical perspective. Exp Cell Res 2007; 313:1981-94. [PMID: 17493611 PMCID: PMC1950476 DOI: 10.1016/j.yexcr.2007.04.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Revised: 04/03/2007] [Accepted: 04/05/2007] [Indexed: 01/08/2023]
Abstract
Intracellular protein filaments intermediate in size between actin microfilaments and microtubules are composed of a surprising variety of tissue specific proteins commonly interconnected with other filamentous systems for mechanical stability and decorated by a variety of proteins that provide specialized functions. The sequence conservation of the coiled-coil, alpha-helical structure responsible for polymerization into individual 10 nm filaments defines the classification of intermediate filament proteins into a large gene family. Individual filaments further assemble into bundles and branched cytoskeletons visible in the light microscope. However, it is the diversity of the variable terminal domains that likely contributes most to different functions. The search for the functions of intermediate filament proteins has led to discoveries of roles in diseases of the skin, heart, muscle, liver, brain, adipose tissues and even premature aging. The diversity of uses of intermediate filaments as structural elements and scaffolds for organizing the distribution of decorating molecules contrasts with other cytoskeletal elements. This review is an attempt to provide some recollection of how such a diverse field emerged and changed over about 30 years.
Collapse
Affiliation(s)
- Robert G Oshima
- Oncodevelopmental Biology Program, Cancer Research Center, The Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
47
|
Jing R, Wilhelmsson U, Goodwill W, Li L, Pan Y, Pekny M, Skalli O. Synemin is expressed in reactive astrocytes in neurotrauma and interacts differentially with vimentin and GFAP intermediate filament networks. J Cell Sci 2007; 120:1267-77. [PMID: 17356066 DOI: 10.1242/jcs.03423] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Immature astrocytes and astrocytoma cells contain synemin and three other intermediate filament (IF) proteins: glial fibrillary acidic protein (GFAP), vimentin and nestin. Here, we show that, after neurotrauma, reactive astrocytes produce synemin and thus propose synemin as a new marker of reactive astrocytes. Comparison of synemin mRNA and protein levels in brain tissues and astrocyte cultures from wild-type, Vim-/- and Gfap-/-Vim-/- mice showed that in the absence of vimentin, synemin protein was undetectable although synemin mRNA was present at wild-type levels. By contrast, in Gfap-/- astrocytes, synemin protein and mRNA levels, as well as synemin incorporation into vimentin IFs, were unaltered. Biochemical assays with purified proteins suggested that synemin interacts with GFAP IFs like an IF-associated protein rather than like a polymerization partner, whereas the opposite was true for synemin interaction with vimentin. In transfection experiments, synemin did not incorporate into normal, filamentous GFAP networks, but integrated into vimentin and GFAP heteropolymeric networks. Thus, alongside GFAP, vimentin and nestin, reactive astrocytes contain synemin, whose accumulation is suppressed post-transcriptionally in the absence of a polymerization partner. In astrocytes, this partner is vimentin and not GFAP, which implies a functional difference between these two type III IF proteins.
Collapse
Affiliation(s)
- Runfeng Jing
- Department of Cellular Biology and Anatomy and Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Blechingberg J, Holm IE, Nielsen KB, Jensen TH, Jørgensen AL, Nielsen AL. Identification and characterization of GFAPkappa, a novel glial fibrillary acidic protein isoform. Glia 2007; 55:497-507. [PMID: 17203480 DOI: 10.1002/glia.20475] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Glial fibrillary acidic protein (GFAP) is the principal component of the intermediary filaments in mature astrocytes of the central nervous system (CNS). The protein consists of three domains: the head, the coiled-coil, and the tail. Here, we describe the isolation of an evolutionary conserved novel GFAP isoform, GFAPkappa, produced by alternative splicing and polyadenylation of the 3'-region of the human GFAP pre-mRNA. As a consequence, the resulting human GFAPkappa protein harbors a nonconserved C-terminal tail sequence distinct from the tails of GFAPalpha, the predominant GFAP isoform, and GFAPepsilon, an isoform which also results from alternative splicing. The head and coiled-coil rod domains are identical between the three GFAP isoforms. Interestingly, GFAPkappa is incapable of forming homomeric filaments, and increasing GFAPkappa expression levels causes a collapse of intermediate filaments formed by GFAPalpha. In searching for a biological relevance of GFAPkappa, we noticed that mRNA expression levels of GFAPalpha, GFAPepsilon, and GFAPkappa are gradually increased during development of the embryonic pig brain. However, whereas the GFAPalpha/GFAPepsilon ratio is constant, the GFAPkappa/GFAPepsilon ratio decreases during brain development. Furthermore, in glioblastoma tumors, an increased GFAPkappa/GFAPepsilon ratio is detected. Our results suggest that the relative expression level of the GFAPkappa isoform could modulate the properties of GFAP intermediate filaments and perhaps thereby influencing the motility of GFAP positive astrocytes and progenitor cells within the CNS.
Collapse
Affiliation(s)
- Jenny Blechingberg
- Institute of Human Genetics, The Bartholin Building, University of Aarhus, Aarhus C DK-8000, Denmark
| | | | | | | | | | | |
Collapse
|
49
|
Izmiryan A, Cheraud Y, Khanamiryan L, Leterrier JF, Federici T, Peltekian E, Moura-Neto V, Paulin D, Li Z, Xue ZG. Different expression of synemin isoforms in glia and neurons during nervous system development. Glia 2006; 54:204-13. [PMID: 16817202 DOI: 10.1002/glia.20378] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The synemin gene encodes proteins belonging to the intermediate filament family. These proteins confer resistance to mechanical stress and modulate cell shape. Three synemin isoforms, of 180 (H), 150 (M) and 41 (L) kDa, are produced by alternative splicing of the pre-mRNA and are regulated differently during development. The three isoforms differ in their C-terminal tail domains, while their IF rod domains are identical. Synemins H/M occurred together with nestin and vimentin in glial progenitors during the early differentiation of the developing mouse central nervous system. They are later found in GFAP-labeled cells. In contrast, the L isoform appeared only in neurons, together with neurofilaments and betaIII-tubulin in the brain after birth. However, synemin L appeared from E13 in the peripheral nervous system, where it was confined to the neurons of spinal ganglia. In the meantime, the synemin H/M isoforms were found in both the neurons and Schwann cells of the sensorial ganglia from E11. Tissue fractionation and purification of IFs from adult mouse spinal cord revealed that the synemin L isoform binds to neurofilaments associated with the membrane compartment. This report describes the synthesis of the three synemin isoforms by selective cell types, and their temporal and spatial distributions. Mechanisms specific to neurons and glia probably control the splicing of the common synemin mRNA and the synthesis of each synemin isoform.
Collapse
Affiliation(s)
- A Izmiryan
- Université Pierre et Marie Curie-Paris 6, UMR 7079, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Davies KE, Nowak KJ. Molecular mechanisms of muscular dystrophies: old and new players. Nat Rev Mol Cell Biol 2006; 7:762-73. [PMID: 16971897 DOI: 10.1038/nrm2024] [Citation(s) in RCA: 224] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The study of the muscle cell in the muscular dystrophies (MDs) has shown that mutant proteins result in perturbations of many cellular components. MDs have been associated with mutations in structural proteins, signalling molecules and enzymes as well as mutations that result in aberrant processing of mRNA or alterations in post-translational modifications of proteins. These findings have not only revealed important insights for cell biologists, but have also provided unexpected and exciting new approaches for therapy.
Collapse
Affiliation(s)
- Kay E Davies
- Department of Physiology, Anatomy and Genetics, MRC Functional Genetics Unit, South Parks Road, Oxford OX1 3QX, UK.
| | | |
Collapse
|