Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Pathol ; 184(8): 2322-32, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24907641

ABSTRACT

Myotonic dystrophy type 2 (DM2) is a multisystemic disorder caused by a (CCTG)n repeat expansion in intron 1 of CNBP. Transcription of the repeats causes a toxic RNA gain of function involving their accumulation in ribonuclear foci. This leads to sequestration of splicing factors and alters pre-mRNA splicing in a range of downstream effector genes, which is thought to contribute to the diverse DM2 clinical features. Hyperlipidemia is frequent in DM2 patients, but the treatment is problematic because of an increased risk of statin-induced adverse reactions. Hypothesizing that shared pathways lead to the increased risk, we compared the skeletal muscle expression profiles of DM2 patients and controls with patients with hyperlipidemia on statin therapy. Neural precursor cell expressed, developmentally downregulated-4 (NEDD4), an ubiquitin ligase, was one of the dysregulated genes identified in DM2 patients and patients with statin-treated hyperlipidemia. In DM2 muscle, NEDD4 mRNA was abnormally spliced, leading to aberrant NEDD4 proteins. NEDD4 was down-regulated in persons taking statins, and simvastatin treatment of C2C12 cells suppressed NEDD4 transcription. Phosphatase and tensin homologue (PTEN), an established NEDD4 target, was increased and accumulated in highly atrophic DM2 muscle fibers. PTEN ubiquitination was reduced in DM2 myofibers, suggesting that the NEDD4-PTEN pathway is dysregulated in DM2 skeletal muscle. Thus, this pathway may contribute to the increased risk of statin-adverse reactions in patients with DM2.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle, Skeletal/metabolism , Myotonic Dystrophy/metabolism , Ubiquitin-Protein Ligases/metabolism , Adult , Blotting, Western , Endosomal Sorting Complexes Required for Transport/genetics , Female , Fluorescent Antibody Technique , Genotype , Humans , Hyperlipidemias/drug therapy , Hyperlipidemias/etiology , Immunohistochemistry , Liver-Specific Organic Anion Transporter 1 , Male , Middle Aged , Myotonic Dystrophy/complications , Myotonic Dystrophy/genetics , Nedd4 Ubiquitin Protein Ligases , Oligonucleotide Array Sequence Analysis , Organic Anion Transporters/genetics , PTEN Phosphohydrolase/metabolism , RNA Splicing , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transcriptome , Ubiquitin-Protein Ligases/genetics
2.
PLoS One ; 9(3): e90819, 2014.
Article in English | MEDLINE | ID: mdl-24618559

ABSTRACT

Tibial muscular dystrophy (TMD) is a late onset, autosomal dominant distal myopathy that results from mutations in the two last domains of titin. The cascade of molecular events leading from the causative Titin mutations to the preterm death of muscle cells in TMD is largely unknown. In this study we examined the mRNA and protein changes associated with the myopathology of TMD. To identify these components we performed gene expression profiling using muscle biopsies from TMD patients and healthy controls. The profiling results were confirmed through quantitative real-time PCR and protein level analysis. One of the pathways identified was activation of endoplasmic reticulum (ER) stress response. ER stress activates the unfolded protein response (UPR) pathway. UPR activation was supported by elevation of the marker genes HSPA5, ERN1 and the UPR specific XBP1 splice form. However, UPR activation appears to be insufficient to correct the protein abnormalities causing its activation because degenerative TMD muscle fibres show an increase in ubiquitinated protein inclusions. Abnormalities of VCP-associated degradation pathways are also suggested by the presence of proteolytic VCP fragments in western blotting, and VCP's accumulation within rimmed vacuoles in TMD muscle fibres together with p62 and LC3B positive autophagosomes. Thus, pathways controlling turnover and degradation, including autophagy, are distorted and lead to degeneration and loss of muscle fibres.


Subject(s)
Autophagy/genetics , Distal Myopathies/genetics , Distal Myopathies/metabolism , Gene Expression Profiling , Transcriptome , Unfolded Protein Response , Adult , Aged , Aged, 80 and over , Biopsy , Case-Control Studies , Cluster Analysis , Distal Myopathies/pathology , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum-Associated Degradation , Female , Humans , Male , Middle Aged , Muscles/metabolism , Muscles/pathology , Signal Transduction
3.
Ann Neurol ; 75(2): 230-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24395473

ABSTRACT

OBJECTIVE: Several patients with previously reported titin gene (TTN) mutations causing tibial muscular dystrophy (TMD) have more complex, severe, or unusual phenotypes. This study aimed to clarify the molecular cause of the variant phenotypes in 8 patients of 7 European families. METHODS: Clinical, histopathological, and muscle imaging data of patients and family members were reanalyzed. The titin protein was analyzed by Western blotting and TTN gene by reverse transcription polymerase chain reaction (RT-PCR) and Sanger sequencing. RESULTS: Western blotting showed more pronounced C-terminal titin abnormality than expected for heterozygous probands, suggesting the existence of additional TTN mutations. RT-PCR indicated unequal mRNA expression of the TTN alleles in biopsies of 6 patients, 3 with an limb-girdle muscular dystrophy type 2J (LGMD2J) phenotype. Novel frameshift mutations were identified in 5 patients. A novel A-band titin mutation, c.92167C>T (p.P30723S), was found in 1 patient, and 1 Portuguese patient with a severe TMD phenotype proved to be homozygous for the previously reported Iberian TMD mutation. INTERPRETATION: The unequal expression levels of TTN transcripts in 5 probands suggested severely reduced expression of the frameshift mutated allele, probably through nonsense-mediated decay, explaining the more severe phenotypes. The Iberian TMD mutation may cause a more severe TMD rather than LGMD2J when homozygous. The Finnish patient compound heterozygous for the FINmaj TMD mutation and the novel A-band titin missense mutation showed a phenotype completely different from previously described titinopathies. Our results further expand the complexity of muscular dystrophies caused by TTN mutations and suggest that the coexistence of second mutations may constitute a more common general mechanism explaining phenotype variability.


Subject(s)
Connectin/genetics , Distal Myopathies/genetics , Distal Myopathies/pathology , Mutation/genetics , Adolescent , Adult , Aged , Base Sequence , Exons/genetics , Exons/immunology , Family Health , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Muscle, Skeletal/pathology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , White People
4.
Ann Neurol ; 73(4): 500-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23401021

ABSTRACT

OBJECTIVE: A study was undertaken to identify the molecular cause of Welander distal myopathy (WDM), a classic autosomal dominant distal myopathy. METHODS: The genetic linkage was confirmed and defined by microsatellite and single nucleotide polymorphism haplotyping. The whole linked genomic region was sequenced with targeted high-throughput and Sanger sequencing, and coding transcripts were sequenced on the cDNA level. WDM muscle biopsies were studied by Western blotting and immunofluorescence microscopy. Splicing of TIA1 and its target genes in muscle and myoblast cultures was analyzed by reverse transcriptase polymerase chain reaction. Mutant TIA1 was characterized by cell biological studies on HeLa cells, including quantification of stress granules by high content analysis and fluorescence recovery after photobleaching (FRAP) experiments. RESULTS: The linked haplotype at 2p13 was narrowed down to <806 kb. Sequencing by multiple methods revealed only 1 segregating coding mutation, c.1362 G>A (p.E384K) in the RNA-binding protein TIA1, a key component of stress granules. Immunofluorescence microscopy of WDM biopsies showed a focal increase of TIA1 in atrophic and vacuolated fibers. In HeLa cells, mutant TIA1 constructs caused a mild increase in stress granule abundance compared to wild type, and showed slower average fluorescence recovery in FRAP. INTERPRETATION: WDM is caused by mutated TIA1 through a dominant pathomechanism probably involving altered stress granule dynamics.


Subject(s)
Distal Myopathies/genetics , Genetic Predisposition to Disease/genetics , Mutation/genetics , Poly(A)-Binding Proteins/genetics , Cells, Cultured , Female , Fluorescence Recovery After Photobleaching , Genetic Linkage , Genotype , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Male , Microsatellite Repeats/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Photobleaching , Polymorphism, Single Nucleotide/genetics , Proteins/genetics , Proteins/metabolism , T-Cell Intracellular Antigen-1 , Ubiquitin/metabolism
5.
Nat Genet ; 44(4): 450-5, S1-2, 2012 Feb 26.
Article in English | MEDLINE | ID: mdl-22366786

ABSTRACT

Limb-girdle muscular dystrophy type 1D (LGMD1D) was linked to chromosome 7q36 over a decade ago, but its genetic cause has remained elusive. Here we studied nine LGMD-affected families from Finland, the United States and Italy and identified four dominant missense mutations leading to p.Phe93Leu or p.Phe89Ile changes in the ubiquitously expressed co-chaperone DNAJB6. Functional testing in vivo showed that the mutations have a dominant toxic effect mediated specifically by the cytoplasmic isoform of DNAJB6. In vitro studies demonstrated that the mutations increase the half-life of DNAJB6, extending this effect to the wild-type protein, and reduce its protective anti-aggregation effect. Further, we show that DNAJB6 interacts with members of the CASA complex, including the myofibrillar myopathy-causing protein BAG3. Our data identify the genetic cause of LGMD1D, suggest that its pathogenesis is mediated by defective chaperone function and highlight how mutations in a ubiquitously expressed gene can exert effects in a tissue-, isoform- and cellular compartment-specific manner.


Subject(s)
HSP40 Heat-Shock Proteins/genetics , Molecular Chaperones/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/metabolism , Nerve Tissue Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins , Finland , Genotype , HSP40 Heat-Shock Proteins/metabolism , Humans , Italy , Molecular Chaperones/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/pathology , Mutation, Missense , Nerve Tissue Proteins/metabolism , United States , Zebrafish/embryology , Zebrafish/genetics
6.
Development ; 135(19): 3311-20, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18776147

ABSTRACT

Trophoblast giant cells are instrumental in promoting blood flow towards the mouse embryo by invading the uterine endometrium and remodelling the maternal vasculature. This process involves the degradation of the perivascular smooth muscle layer and the displacement of vascular endothelial cells to form trophoblast-lined blood sinuses. How this vascular remodelling is achieved at the molecular level remains largely elusive. Here, we show that two placenta-specific cathepsins, Cts7 and Cts8, are expressed in distinct but largely overlapping subsets of giant cells that are in direct contact with maternal arteries. We find that Cts8, but not Cts7, has the capacity to mediate loss of smooth muscle alpha-actin and to disintegrate blood vessels. Consequently, conditional ubiquitous overexpression of Cts8 leads to midgestational embryonic lethality caused by severe vascularization defects. In addition, both cathepsins determine trophoblast cell fate by inhibiting the self-renewing capacity of trophoblast stem cells when overexpressed in vitro. Similarly, transgenic overexpression of Cts7 and Cts8 affects trophoblast proliferation and differentiation by prolonging mitotic cell cycle progression and promoting giant cell differentiation, respectively. We also show that the cell cycle effect is directly caused by some proportion of CTS7 localizing to the nucleus, highlighting the emerging functional diversity of these typically lysosomal proteases in distinct intracellular compartments. Our findings provide evidence for the highly specialized functions of closely related cysteine cathepsin proteases in extra-embryonic development, and reinforce their importance for a successful outcome of pregnancy.


Subject(s)
Cathepsins/physiology , Placenta/blood supply , Placenta/enzymology , Trophoblasts/physiology , Animals , Cathepsins/genetics , Cell Cycle , Cell Differentiation , Cell Proliferation , Embryonic Development/genetics , Endosomes/enzymology , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Giant Cells/cytology , Giant Cells/enzymology , Lysosomes/enzymology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Models, Biological , Pregnancy , Proteins/genetics , Proteins/physiology , Trophoblasts/cytology
SELECTION OF CITATIONS
SEARCH DETAIL