Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Biol Chem ; 300(5): 107254, 2024 May.
Article in English | MEDLINE | ID: mdl-38569934

ABSTRACT

Nesprins comprise a family of multi-isomeric scaffolding proteins, forming the linker of nucleoskeleton-and-cytoskeleton complex with lamin A/C, emerin and SUN1/2 at the nuclear envelope. Mutations in nesprin-1/-2 are associated with Emery-Dreifuss muscular dystrophy (EDMD) with conduction defects and dilated cardiomyopathy (DCM). We have previously observed sarcomeric staining of nesprin-1/-2 in cardiac and skeletal muscle, but nesprin function in this compartment remains unknown. In this study, we show that specific nesprin-2 isoforms are highly expressed in cardiac muscle and localize to the Z-disc and I band of the sarcomere. Expression of GFP-tagged nesprin-2 giant spectrin repeats 52 to 53, localized to the sarcomere of neonatal rat cardiomyocytes. Yeast two-hybrid screening of a cardiac muscle cDNA library identified telethonin and four-and-half LIM domain (FHL)-2 as potential nesprin-2 binding partners. GST pull-down and immunoprecipitation confirmed the individual interactions between nesprin-2/telethonin and nesprin-2/FHL-2, and showed that nesprin-2 and telethonin binding was dependent on telethonin phosphorylation status. Importantly, the interactions between these binding partners were impaired by mutations in nesprin-2, telethonin, and FHL-2 identified in EDMD with DCM and hypertrophic cardiomyopathy patients. These data suggest that nesprin-2 is a novel sarcomeric scaffold protein that may potentially participate in the maintenance and/or regulation of sarcomeric organization and function.


Subject(s)
Connectin , LIM Domain Proteins , Muscle Proteins , Myocytes, Cardiac , Nerve Tissue Proteins , Nuclear Proteins , Sarcomeres , Animals , Humans , Mice , Rats , Connectin/metabolism , Connectin/genetics , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , LIM-Homeodomain Proteins , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Muscle Proteins/metabolism , Muscle Proteins/genetics , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Protein Binding , Sarcomeres/metabolism , Transcription Factors
2.
EBioMedicine ; 51: 102587, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31862442

ABSTRACT

BACKGROUND: As genome-wide approaches prove difficult with genetically heterogeneous orphan diseases, we developed a new approach to identify candidate genes. We applied this to Emery-Dreifuss muscular dystrophy (EDMD), characterised by early onset contractures, slowly progressive muscular wasting, and life-threatening heart conduction disturbances with wide intra- and inter-familial clinical variability. Roughly half of EDMD patients are linked to six genes encoding nuclear envelope proteins, but the disease mechanism remains unclear because the affected proteins function in both cell mechanics and genome regulation. METHODS: A primer library was generated to test for mutations in 301 genes from four categories: (I) all known EDMD-linked genes; (II) genes mutated in related muscular dystrophies; (III) candidates generated by exome sequencing in five families; (IV) functional candidates - other muscle nuclear envelope proteins functioning in mechanical/genome processes affected in EDMD. This was used to sequence 56 unlinked patients with EDMD-like phenotype. FINDINGS: Twenty-one patients could be clearly assigned: 18 with mutations in genes of similar muscular dystrophies; 3 with previously missed mutations in EDMD-linked genes. The other categories yielded novel candidate genes, most encoding nuclear envelope proteins with functions in gene regulation. INTERPRETATION: Our multi-pronged approach identified new disease alleles and many new candidate EDMD genes. Their known functions strongly argue the EDMD pathomechanism is from altered gene regulation and mechanotransduction due to connectivity of candidates from the nuclear envelope to the plasma membrane. This approach highlights the value of testing for related diseases using primer libraries and may be applied for other genetically heterogeneous orphan diseases. FUNDING: The Wellcome Trust, Muscular Dystrophy UK, Medical Research Council, European Community's Seventh Framework Programme "Integrated European -omics research project for diagnosis and therapy in rare neuromuscular and neurodegenerative diseases (NEUROMICS)".


Subject(s)
Alleles , Gene Expression Regulation , Muscular Dystrophy, Emery-Dreifuss/genetics , Sequence Analysis, DNA , Gene Ontology , Muscles/metabolism , Mutation/genetics , Exome Sequencing
3.
Neuromuscul Disord ; 27(4): 338-351, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28214269

ABSTRACT

Reports of aberrant distribution for some nuclear envelope proteins in cells expressing a few Emery-Dreifuss muscular dystrophy mutations raised the possibility that such protein redistribution could underlie pathology and/or be diagnostic. However, this disorder is linked to 8 different genes encoding nuclear envelope proteins, raising the question of whether a particular protein is most relevant. Therefore, myoblast/fibroblast cultures from biopsy and tissue sections from a panel of nine Emery-Dreifuss muscular dystrophy patients (4 male, 5 female) including those carrying emerin and FHL1 (X-linked) and several lamin A (autosomal dominant) mutations were stained for the proteins linked to the disorder. As tissue-specific nuclear envelope proteins have been postulated to mediate the tissue-specific pathologies of different nuclear envelopathies, patient samples were also stained for several muscle-specific nuclear membrane proteins. Although linked proteins nesprin 1 and SUN2 and muscle-specific proteins NET5/Samp1 and Tmem214 yielded aberrant distributions in individual patient cells, none exhibited defects through the larger patient panel. Muscle-specific Tmem38A normally appeared in both the nuclear envelope and sarcoplasmic reticulum, but most patient samples exhibited a moderate redistribution favouring the sarcoplasmic reticulum. The absence of striking uniform defects in nuclear envelope protein distribution indicates that such staining will be unavailing for general diagnostics, though it remains possible that specific mutations exhibiting protein distribution defects might reflect a particular clinical variant. These findings further argue that multiple pathways can lead to the generally similar pathologies of this disorder while at the same time the different cellular phenotypes observed possibly may help explain the considerable clinical variation of EDMD.


Subject(s)
Muscular Dystrophy, Emery-Dreifuss/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Tissue Banks , Adolescent , Adult , Biomarkers/metabolism , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Male , Middle Aged , Muscle, Skeletal , Muscular Dystrophy, Emery-Dreifuss/pathology
4.
Nucleus ; 6(1): 66-76, 2015.
Article in English | MEDLINE | ID: mdl-25738644

ABSTRACT

More than 20 mutations in the gene encoding A-type lamins (LMNA) cause progeria, a rare premature aging disorder. The major pathognomonic hallmarks of progeria cells are seen as nuclear deformations or blebs that are related to the redistribution of A- and B-type lamins within the nuclear lamina. However, the functional significance of these progeria-associated blebs remains unknown. We have carried out an analysis of the structural and functional consequences of progeria-associated nuclear blebs in dermal fibroblasts from a progeria patient carrying a rare point mutation p.S143F (C428T) in lamin A/C. These blebs form microdomains that are devoid of major structural components of the nuclear envelope (NE)/lamina including B-type lamins and nuclear pore complexes (NPCs) and are enriched in A-type lamins. Using laser capture microdissection and comparative genomic hybridization (CGH) analyses, we show that, while these domains are devoid of centromeric heterochromatin and gene-poor regions of chromosomes, they are enriched in gene-rich chromosomal regions. The active form of RNA polymerase II is also greatly enriched in blebs as well as nascent RNA but the nuclear co-activator SKIP is significantly reduced in blebs compared to other transcription factors. Our results suggest that the p.S143F progeria mutation has a severe impact not only on the structure of the lamina but also on the organization of interphase chromatin domains and transcription. These structural defects are likely to contribute to gene expression changes reported in progeria and other types of laminopathies.


Subject(s)
Cell Nucleus/genetics , Chromosomes, Human/genetics , Lamin Type B/deficiency , Progeria/genetics , Progeria/pathology , Cell Nucleus/metabolism , Child , Female , Humans , Lamin Type B/genetics , Mutation , Transcription, Genetic
5.
Neuromuscul Disord ; 25(2): 127-36, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25454731

ABSTRACT

Emery-Dreifuss muscular dystrophy (EDMD) is a neuromuscular disease characterized by early contractures, slowly progressive muscular weakness and life-threatening cardiac arrhythmia that can develop into cardiomyopathy. In X-linked EDMD (EDMD1), female carriers are usually unaffected. Here we present a clinical description and in vitro characterization of a mildly affected EDMD1 female carrying the heterozygous EMD mutation c.174_175delTT; p.Y59* that yields loss of protein. Muscle tissue sections and cultured patient myoblasts exhibited a mixed population of emerin-positive and -negative cells; thus uneven X-inactivation was excluded as causative. Patient blood cells were predominantly emerin-positive, but considerable nuclear lobulation was observed in non-granulocyte cells - a novel phenotype in EDMD. Both emerin-positive and emerin-negative myoblasts exhibited spontaneous differentiation in tissue culture, though emerin-negative myoblasts were more proliferative than emerin-positive cells. The preferential proliferation of emerin-negative myoblasts together with the high rate of spontaneous differentiation in both populations suggests that loss of functional satellite cells might be one underlying mechanism for disease pathology. This could also account for the slowly developing muscle phenotype.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Muscular Dystrophy, Emery-Dreifuss/pathology , Myoblasts/pathology , Adolescent , Adult , Age of Onset , Antigens, CD , Autoantigens/metabolism , Cardiomyopathies/etiology , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Child , Family Health , Female , Flow Cytometry , Humans , Ki-67 Antigen/metabolism , Lamin Type A/metabolism , Magnetic Resonance Imaging , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscular Dystrophy, Emery-Dreifuss/complications , Muscular Dystrophy, Emery-Dreifuss/genetics , Myoblasts/metabolism , Nuclear Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection , Young Adult
6.
Aging (Albany NY) ; 6(9): 755-70, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25324471

ABSTRACT

Lamin A is a key component of the nuclear lamina produced through post-translational processing of its precursor known as prelamin A.LMNA mutations leading to farnesylated prelamin A accumulation are known to cause lipodystrophy, progeroid and developmental diseases, including Mandibuloacral dysplasia, a mild progeroid syndrome with partial lipodystrophy and altered bone turnover. Thus, degradation of prelamin A is expected to improve the disease phenotype. Here, we show different susceptibilities of prelamin A forms to proteolysis and further demonstrate that treatment with rapamycin efficiently and selectively triggers lysosomal degradation of farnesylated prelamin A, the most toxic processing intermediate. Importantly, rapamycin treatment of Mandibuloacral dysplasia cells, which feature very low levels of the NAD-dependent sirtuin SIRT-1 in the nuclear matrix, restores SIRT-1 localization and distribution of chromatin markers, elicits release of the transcription factor Oct-1 and determines shortening of the prolonged S-phase. These findings indicate the drug as a possible treatment for Mandibuloacral dysplasia.


Subject(s)
Acro-Osteolysis/drug therapy , Antibiotics, Antineoplastic/therapeutic use , Lipodystrophy/drug therapy , Mandible/abnormalities , Nuclear Proteins/metabolism , Octamer Transcription Factor-1/metabolism , Protein Precursors/metabolism , Sirolimus/therapeutic use , Acro-Osteolysis/metabolism , Adult , Antibiotics, Antineoplastic/pharmacology , Cell Cycle/drug effects , Cells, Cultured , Chromatin/drug effects , Contracture/metabolism , DNA Repair/drug effects , DNA-Binding Proteins/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Infant, Newborn , Lamin Type A , Lipodystrophy/metabolism , Mandible/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/genetics , Protein Precursors/genetics , Sirolimus/pharmacology , Skin Abnormalities/metabolism
7.
PLoS Genet ; 10(9): e1004605, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25210889

ABSTRACT

Proteins of the nuclear envelope (NE) are associated with a range of inherited disorders, most commonly involving muscular dystrophy and cardiomyopathy, as exemplified by Emery-Dreifuss muscular dystrophy (EDMD). EDMD is both genetically and phenotypically variable, and some evidence of modifier genes has been reported. Six genes have so far been linked to EDMD, four encoding proteins associated with the LINC complex that connects the nucleus to the cytoskeleton. However, 50% of patients have no identifiable mutations in these genes. Using a candidate approach, we have identified putative disease-causing variants in the SUN1 and SUN2 genes, also encoding LINC complex components, in patients with EDMD and related myopathies. Our data also suggest that SUN1 and SUN2 can act as disease modifier genes in individuals with co-segregating mutations in other EDMD genes. Five SUN1/SUN2 variants examined impaired rearward nuclear repositioning in fibroblasts, confirming defective LINC complex function in nuclear-cytoskeletal coupling. Furthermore, myotubes from a patient carrying compound heterozygous SUN1 mutations displayed gross defects in myonuclear organization. This was accompanied by loss of recruitment of centrosomal marker, pericentrin, to the NE and impaired microtubule nucleation at the NE, events that are required for correct myonuclear arrangement. These defects were recapitulated in C2C12 myotubes expressing exogenous SUN1 variants, demonstrating a direct link between SUN1 mutation and impairment of nuclear-microtubule coupling and myonuclear positioning. Our findings strongly support an important role for SUN1 and SUN2 in muscle disease pathogenesis and support the hypothesis that defects in the LINC complex contribute to disease pathology through disruption of nuclear-microtubule association, resulting in defective myonuclear positioning.


Subject(s)
Cell Nucleus/metabolism , Cytoskeleton/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Microtubule-Associated Proteins/genetics , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Nuclear Proteins/genetics , Animals , Cell Nucleus/genetics , Cells, Cultured , Cytoskeleton/genetics , Cytoskeleton/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Dystrophies/metabolism , Mutation/genetics , Myoblasts/metabolism , Myoblasts/pathology , NIH 3T3 Cells , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Envelope/pathology , Nuclear Proteins/metabolism
8.
Eur J Hum Genet ; 22(8): 1002-11, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24169522

ABSTRACT

Restrictive dermopathy (RD) is a rare and extremely severe congenital genodermatosis, characterized by a tight rigid skin with erosions at flexure sites, multiple joint contractures, low bone density and pulmonary insufficiency generally leading to death in the perinatal period. RD is caused in most patients by compound heterozygous or homozygous ZMPSTE24 null mutations. This gene encodes a metalloprotease specifically involved in lamin A post-translational processing. Here, we report a total of 16 families for whom diagnosis and molecular defects were clearly established. Among them, we report seven new ZMPSTE24 mutations, identified in classical RD or Mandibulo-acral dysplasia (MAD) affected patients. We also report nine families with one or two affected children carrying the common, homozygous thymine insertion in exon 9 and demonstrate the lack of a founder effect. In addition, we describe several new ZMPSTE24 variants identified in unaffected controls or in patients affected with non-classical progeroid syndromes. In addition, this mutation update includes a comprehensive search of the literature on previously described ZMPSTE24 mutations and associated phenotypes. Our comprehensive analysis of the molecular pathology supported the general rule: complete loss-of-function of ZMPSTE24 leads to RD, whereas other less severe phenotypes are associated with at least one haploinsufficient allele.


Subject(s)
Contracture/genetics , Fetal Growth Retardation/genetics , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mutation , Progeria/genetics , Skin Abnormalities/genetics , Alleles , Amino Acid Substitution , Contracture/diagnosis , DNA Mutational Analysis , Exons , Female , Fetal Growth Retardation/diagnosis , Founder Effect , Genetic Association Studies , Humans , Introns , Male , Pedigree , Progeria/diagnosis , RNA Splice Sites , Skin Abnormalities/diagnosis
9.
Hum Mutat ; 35(4): 452-61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24375709

ABSTRACT

Mutations in several genes encoding nuclear envelope (NE) associated proteins cause Emery-Dreifuss muscular dystrophy (EDMD). We analyzed fibroblasts from a patient who had a mutation in the EMD gene (p.L84Pfs*6) leading to loss of Emerin and a heterozygous mutation in SUN1 (p.A203V). The second patient harbored a heterozygous mutation in LAP2alpha (p.P426L) and a further mutation in SUN1 (p.A614V). p.A203V is located in the N-terminal domain of SUN1 facing the nucleoplasm and situated in the vicinity of the Nesprin-2 and Emerin binding site. p.A614V precedes the SUN domain, which interacts with the KASH domain of Nesprins in the periplasmic space and forms the center of the LINC complex. At the cellular level, we observed alterations in the amounts for several components of the NE in patient fibroblasts and further phenotypic characteristics generally attributed to laminopathies such as increased sensitivity to heat stress. The defects were more severe than observed in EDMD cells with mutations in a single gene. In particular, in patient fibroblasts carrying the p.A203V mutation in SUN1, the alterations were aggravated. Moreover, SUN1 of both patient fibroblasts exhibited reduced interaction with Lamin A/C and when expressed ectopically in wild-type fibroblasts, the SUN1 mutant proteins exhibited reduced interactions with Emerin as well.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Thymopoietins/genetics , DNA-Binding Proteins/genetics , Female , Fibroblasts/metabolism , Humans , Lamin Type A/metabolism , Male , Mutation , Nuclear Envelope/metabolism , Thymopoietins/metabolism
10.
Aging (Albany NY) ; 5(6): 445-59, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23804595

ABSTRACT

The clinical course of a female patient affected by a progeroid syndrome with Restrictive Dermopathy (RD)-like features was followed up. Besides missing hairiness, stagnating weight and growth, RD-like features including progressive skin swelling and solidification, acrocontractures, osteolysis and muscular hypotension were observed until the patient died at the age of 11 months. A homozygousLMNA mutation c.1303C>T (p.R435C) was found by Sanger sequencing. Haplotyping revealed a partial uniparental disomy of chromosome 1 (1q21.3 to 1q23.1) including the LMNA gene. In contrast to reported RD patients with LMNA mutations, LMNA p.R435C is not located at the cleavage site necessary for processing of prelamin A by ZMPSTE24 and leads to a distinct phenotype combining clinical features of Restrictive Dermopathy, Mandibuloacral Dysplasia and Hutchinson-Gilford Progeria. Functionally, LMNA p.R435C is associated with increasing DNA double strand breaks and decreased recruitment of P53 binding protein 1 (53BP1) to DNA-damage sites indicating delayed DNA repair. The follow-up of the complete clinical course in the patient combined with functional studies showed for the first time that a progressive loss of lamin A rather than abnormal accumulation of prelamin A species could be a pathophysiological mechanism in progeroid laminopathies, which leads to DNA repair deficiency accompanied by advancing tissue degeneration.


Subject(s)
Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , Lamin Type A/metabolism , Skin Abnormalities/genetics , Skin Abnormalities/pathology , DNA Repair , Fatal Outcome , Female , Humans , Infant , Lamin Type A/genetics , Mutation , Pedigree
11.
Neuromuscul Disord ; 23(7): 587-90, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23746545

ABSTRACT

Primary laminopathies caused by mutations in the LMNA gene typically display an extremely pleiotropic clinical presentation including cardiac, muscular and metabolic phenotypes. Additionally, many atypical laminopathies have been described combining features of two or more of the distinctive disorders or syndromes associated with LMNA mutations. We report on a 46-year-old female patient with a heterozygous p.R28W LMNA mutation, who presented with a novel clinical phenotype comprising severe limb-girdle muscular dystrophy, pronounced partial lipodystrophy, cardiac conduction defect, polycystic ovary disease and a metabolic syndrome with insulin-resistant diabetes mellitus and hypertriglyceridemia. On examination, her 23-year old daughter solely showed early signs of a LGMD phenotype.


Subject(s)
Lamin Type A/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Mutation/genetics , Arrhythmias, Cardiac/genetics , Brugada Syndrome , Cardiac Conduction System Disease , Female , Genetic Predisposition to Disease , Heart Conduction System/abnormalities , Heterozygote , Humans , Metabolic Syndrome/complications , Middle Aged , Muscular Dystrophies, Limb-Girdle/complications , Muscular Dystrophies, Limb-Girdle/diagnosis , Phenotype , Young Adult
12.
Cell Cycle ; 11(19): 3568-77, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22935701

ABSTRACT

Prelamin A processing impairment is a common feature of a restricted group of rare genetic alterations/disorders associated with a wide range of clinical phenotypes. Changes in histone posttranslational modifications, alterations in non-histone chromatin proteins and chromatin disorganization have been specifically linked to impairment of specific, distinct prelamin A processing steps, but the molecular mechanism involved in these processes is not yet understood . In this study, we show that the accumulation of wild-type prelamin A detected in restrictive dermopathy (RD), as well as the accumulation of mutated forms of prelamin A identified in familial partial lipodystrophy (FPLD) and mandibuloacral dysplasia (MADA), affect the nuclear localization of barrier-to-autointegration factor (BAF), a protein able to link lamin A precursor to chromatin remodeling functions. Our findings, in accordance with previously described results, support the hypothesis of a prelamin A involvement in BAF nuclear recruitment and suggest BAF-prelamin A complex as a protein platform usually activated in prelamin A-accumulating diseases. Finally, we demonstrate the involvement of the inner nuclear membrane protein emerin in the proper localization of BAF-prelamin A complex.


Subject(s)
Acro-Osteolysis/metabolism , Cell Nucleus/metabolism , Contracture/metabolism , DNA-Binding Proteins/metabolism , Lipodystrophy, Familial Partial/metabolism , Lipodystrophy/metabolism , Nuclear Proteins/metabolism , Skin Abnormalities/metabolism , Acro-Osteolysis/pathology , Adult , Animals , Contracture/pathology , HEK293 Cells , Humans , Infant, Newborn , Lamin Type A , Lipodystrophy/pathology , Lipodystrophy, Familial Partial/pathology , Mandible/abnormalities , Mandible/metabolism , Mandible/pathology , Membrane Proteins/metabolism , Mutant Proteins/metabolism , Protein Binding , Protein Precursors/metabolism , Protein Transport , Rats , Skin Abnormalities/pathology , Transfection
13.
Eur J Cell Biol ; 91(8): 614-28, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22555292

ABSTRACT

Emery-Dreifuss muscular dystrophy (EDMD) is a late onset-disease characterized by skeletal muscle wasting and heart defects with associated risk of sudden death. The autosomal dominant form of the disease is caused by mutations in the LMNA gene encoding LaminA and C, the X-linked form results from mutations in the gene encoding the inner nuclear membrane protein Emerin (STA). Both Emerin and LaminA/C interact with the nuclear envelope proteins Nesprin-1 and -2 and mutations in genes encoding C-terminal isoforms of Nesprin-1 and -2 have also been implicated in EDMD. Here we analyse primary fibroblasts from patients affected by either Duchenne muscular dystrophy (DMD) or Emery-Dreifuss muscular dystrophy/Charcot-Marie-Tooth syndrome (EDMD/CMT) that in addition to the disease causing mutations harbour mutations in the Nesprin-1 gene and in the SUN1 and SUN2 gene, respectively. SUN proteins together with the Nesprins form the core of the LINC complex which connects the nucleus with the cytoskeleton. The mutations are accompanied by changes in cell adhesion, cell migration, senescence, and stress response, as well as in nuclear shape and nuclear envelope composition which are changes characteristic for laminopathies. Our results point to a potential influence of mutations in components of the LINC complex on the clinical outcome and the molecular pathology in the patients.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Fibroblasts/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Emery-Dreifuss/genetics , Cell Adhesion , Cell Movement , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Nucleus Shape , Cellular Senescence , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Cytoskeletal Proteins , Female , Fibroblasts/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lamin Type A/genetics , Lamin Type A/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Emery-Dreifuss/metabolism , Muscular Dystrophy, Emery-Dreifuss/pathology , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Primary Cell Culture , Stress, Physiological , Transfection , Wound Healing
14.
Biochem Soc Trans ; 39(6): 1693-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22103509

ABSTRACT

The LINC (linker of nucleoskeleton and cytoskeleton) complex is a proposed mechanical link tethering the nucleo- and cyto-skeleton via the NE (nuclear envelope). The LINC components emerin, lamin A/C, SUN1, SUN2, nesprin-1 and nesprin-2 interact with each other at the NE and also with other binding partners including actin filaments and B-type lamins. Besides the mechanostructural functions, the LINC complex is also involved in signalling pathways and gene regulation. Emerin was the first LINC component associated with a human disease, namely EDMD (Emery-Dreifuss muscular dystrophy). Later on, other components of the LINC complex, such as lamins A/C and small isoforms of nesprin-1 and nesprin-2, were found to be associated with EDMD, reflecting a genetic heterogeneity that has not been resolved so far. Only approximately 46% of the EDMD patients can be linked to genes of LINC and non-LINC components, pointing to further genes involved in the pathology of EDMD. Obvious candidates are the LINC proteins SUN1 and SUN2. Recently, screening of binding partners of LINC components as candidates identified LUMA (TMEM43), encoding a binding partner of emerin and lamins, as a gene involved in atypical EDMD. Nevertheless, such mutations contribute only to a very small fraction of EDMD patients. EDMD-causing mutations in STA/EMD (encoding emerin) that disrupt emerin binding to Btf (Bcl-2-associated transcription factor), GCL (germ cell-less) and BAF (barrier to autointegration factor) provide the first glimpses into LINC being involved in gene regulation and thus opening new avenues for functional studies. Thus the association of LINC with human disease provides tools for understanding its functions within the cell.


Subject(s)
Disease , Multiprotein Complexes/metabolism , Base Sequence , Humans , Lamin Type A/genetics , Molecular Sequence Data , Muscular Dystrophy, Emery-Dreifuss/metabolism
15.
Nucleus ; 2(5): 500-9, 2011.
Article in English | MEDLINE | ID: mdl-21983083

ABSTRACT

The nuclear envelope (NE) is a double membrane physical barrier, which separates the nucleus from the cytoplasm. Underlying the NE are the nuclear lamins, which in combination with inner nuclear membrane proteins form the lamina. The lamina is crucial for maintaining the structural integrity of the nucleus and for positioning of nuclear pore complexes (NPCs) within the NE. The nucleoporin Nup153 has previously been reported to bind to B-type lamins. However, the specificity of this interaction is not well established. Here we show that Nup153 exhibits multiple binding sites for A- and B-type lamins. Using GST-pull down assays, we found that both the N-terminal domain of Nup153 and its C terminus associate with the Ig-fold domain of A- and B-type lamins. By employing purified Nup153 and lamin proteins in blot overlay assays we revealed that both the N-terminal and the C-terminal domain of Nup153 are directly interacting with the lamins. Moreover, we provide evidence that mutations in the lamin A Ig-fold domain selectively affect Nup153-binding, suggesting that Nup153 may play a role in lamin-associated diseases, known as laminopathies. Together our results indicate a far more intricate interplay between Nup153 and nuclear lamins than previously accepted.


Subject(s)
Lamin Type A/metabolism , Lamin Type B/metabolism , Nuclear Pore Complex Proteins/metabolism , Binding Sites , Cells, Cultured , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Lamin Type A/chemistry , Lamin Type A/genetics , Lamin Type B/chemistry , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
17.
Dev Dyn ; 239(3): 998-1009, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20108321

ABSTRACT

Nesprins are a family of nuclear transmembrane proteins anchored via Sun proteins to the nuclear membrane. Analysis of nesprins during human muscle development revealed an increase in nesprin-1-giant during early myogenesis in vitro. During the transition from immature to mature muscle fibres in vivo, nesprin-2 partly replaced nesprin-1 at the nuclear envelope and short nesprin isoforms became dominant. Sun1 and Sun2 proteins remained unchanged during this fibre maturation. In emerin-negative skin fibroblasts, nesprin-2-giant was relocated from the nuclear envelope to the cytoplasm, not to the endoplasmic reticulum, while nesprin-1 remained at the nuclear envelope. In emerin-negative keratinocytes lacking nesprin-1, nesprin-2 remained at the nuclear envelope. HeLa cell nuclear envelopes lacked nesprin-1, which was the dominant form in myoblasts, while a novel 130-kD nesprin-2 isoform dominated Ntera-2 cells. The results suggest the possibility of isoform-specific and tissue-specific roles for nesprins in nuclear positioning.


Subject(s)
Microfilament Proteins/chemistry , Muscles/embryology , Nerve Tissue Proteins/chemistry , Nuclear Envelope/metabolism , Nuclear Proteins/chemistry , Animals , Antibodies, Monoclonal/chemistry , Cell Nucleus/metabolism , Cytoskeletal Proteins , Fibroblasts/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Isoforms , Telomere-Binding Proteins/metabolism
18.
Am J Hum Genet ; 85(3): 338-53, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19716112

ABSTRACT

Emery-Dreifuss muscular dystrophy (EDMD) is a rare disorder characterized by early joint contractures, muscular dystrophy, and cardiac involvement with conduction defects and arrhythmias. So far, only 35% of EDMD cases are genetically elucidated and associated with EMD or LMNA gene mutations, suggesting the existence of additional major genes. By whole-genome scan, we identified linkage to the Xq26.3 locus containing the FHL1 gene in three informative families belonging to our EMD- and LMNA-negative cohort. Analysis of the FHL1 gene identified seven mutations, in the distal exons of FHL1 in these families, three additional families, and one isolated case, which differently affect the three FHL1 protein isoforms: two missense mutations affecting highly conserved cysteines, one abolishing the termination codon, and four out-of-frame insertions or deletions. The predominant phenotype was characterized by myopathy with scapulo-peroneal and/or axial distribution, as well as joint contractures, and associated with a peculiar cardiac disease characterized by conduction defects, arrhythmias, and hypertrophic cardiomyopathy in all index cases of the seven families. Heterozygous female carriers were either asymptomatic or had cardiac disease and/or mild myopathy. Interestingly, four of the FHL1-mutated male relatives had isolated cardiac disease, and an overt hypertrophic cardiomyopathy was present in two. Expression and functional studies demonstrated that the FHL1 proteins were severely reduced in all tested patients and that this was associated with a severe delay in myotube formation in the two patients for whom myoblasts were available. In conclusion, FHL1 should be considered as a gene associated with the X-linked EDMD phenotype, as well as with hypertrophic cardiomyopathy.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Muscle Proteins/genetics , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation/genetics , Adolescent , Adult , Cardiovascular Diseases/complications , Cell Differentiation , Child , Child, Preschool , Chromosomes, Human, X/genetics , Cohort Studies , DNA Mutational Analysis , Female , Fluorescent Antibody Technique , Genes, X-Linked , Genome-Wide Association Study , Humans , Immunoblotting , LIM Domain Proteins , Lod Score , Lung Diseases/complications , Male , Middle Aged , Muscular Dystrophy, Emery-Dreifuss/complications , Myoblasts/pathology , Pedigree , Protein Isoforms/genetics , Sarcomeres/pathology
19.
Eur J Cell Biol ; 88(10): 593-608, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19589617

ABSTRACT

We have studied myoblasts from a patient with a severe autosomal dominant Emery-Dreifuss muscular dystrophy (AD-EDMD) caused by an arginine 545 to cystein point mutation (p.R545C) in the carboxy-terminal domain of the lamin A/C gene. This mutation has pleiotropic cellular effects on these myoblasts as demonstrated by nuclear structural defects, exhibiting lobulations which increase with cell passages in culture. The organization of both lamin A/C and its inner nuclear membrane partner emerin are altered, eventually showing a honeycomb pattern upon immunofluorescence microscopy. In addition, the distribution of histone H3 trimethylated at lysine 27 and of phosphorylated RNA polymerase II, markers of inactive and active chromatin domains, respectively, are altered suggesting an impact on gene expression. Patient myoblasts also presented a high index of senescence in ex vivo culture. Moreover, our data show for the first time in an AD-EDMD context that the 20S core particle of the proteasome was inactivated. With cell passages, the 20S core protein progressively accumulated into discrete nuclear foci that largely colocalized with promyelocytic leukemia (PML) bodies while p21 accumulated throughout the nuclear compartment. Proteasome inactivation has been linked to normal cellular ageing. Our data indicate that it may also contribute to premature senescence in AD-EDMD patient myoblasts. Finally, when transferred to low-serum medium, patient myoblasts were deficient in ex vivo differentiation, as assessed by the absence of myotube formation and myogenin induction. Altogether, these data suggest that the LMNA mutation p.R545C impairs both proliferation and differentiation capacities of myoblasts as part of the pathogenesis of AD-EDMD.


Subject(s)
Aging/genetics , Cell Differentiation/genetics , Cell Nucleus/metabolism , Lamin Type A/genetics , Muscular Dystrophy, Emery-Dreifuss/genetics , Myoblasts/metabolism , Amino Acid Substitution , Antibodies/metabolism , Bisbenzimidazole/metabolism , Carbocyanines/metabolism , Case-Control Studies , Cell Culture Techniques , Cell Nucleus/pathology , Cell Nucleus/ultrastructure , Cells, Cultured , Cysteine/metabolism , Female , Fluorescein-5-isothiocyanate/metabolism , Fluorescent Dyes/metabolism , Humans , Male , Muscular Dystrophy, Emery-Dreifuss/metabolism , Muscular Dystrophy, Emery-Dreifuss/pathology , Mutation, Missense , Myoblasts/cytology , Myoblasts/ultrastructure , Point Mutation , Xanthenes/metabolism
20.
Biochim Biophys Acta ; 1792(8): 810-21, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19524666

ABSTRACT

In-frame mutations in nuclear lamin A/C lead to a multitude of tissue-specific degenerative diseases known as the 'laminopathies'. Previous studies have demonstrated that lamin A/C-null mouse fibroblasts have defects in cell polarisation, suggesting a role for lamin A/C in nucleo-cytoskeletal-cell surface cross-talk. However, this has not been examined in patient fibroblasts expressing modified forms of lamin A/C. Here, we analysed skin fibroblasts from 3 patients with Emery-Dreifuss muscular dystrophy and from 1 with dilated cardiomyopathy. The emerin-lamin A/C interaction was impaired in each mutant cell line. Mutant cells exhibited enhanced cell proliferation, collagen-dependent adhesion, larger numbers of filopodia and smaller cell spread size, compared with control cells. Furthermore, cell migration, speed and polarization were elevated. Mutant cells also showed an enhanced ability to contract collagen gels at early time points, compared with control cells. Phosphotyrosine measurements during cell spreading indicated an initial temporal lag in ERK1/2 activation in our mutant cells, followed by hyper-activation of ERK1/2 at 2 h post cell attachment. Deregulated ERK1/2 activation is linked with cardiomyopathy, cell spreading and proliferation defects. We conclude that a functional emerin-lamin A/C complex is required for cell spreading and proliferation, possibly acting through ERK1/2 signalling.


Subject(s)
Fibroblasts/physiology , Lamin Type A/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Adolescent , Adult , Animals , Cell Adhesion , Cell Cycle , Cell Movement , Cell Proliferation , Enzyme Activation , Female , Fibroblasts/cytology , Humans , Lamin Type A/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Middle Aged , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...