Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Neuropathol Appl Neurobiol ; 49(1): e12853, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36180966

ABSTRACT

AIMS: Target skeletal muscle fibres - defined by different concentric areas in oxidative enzyme staining - can occur in patients with neurogenic muscular atrophy. Here, we used our established hypothesis-free proteomic approach with the aim of deciphering the protein composition of targets. We also searched for potential novel interactions between target proteins. METHODS: Targets and control areas were laser microdissected from skeletal muscle sections of 20 patients with neurogenic muscular atrophy. Samples were analysed by a highly sensitive mass spectrometry approach, enabling relative protein quantification. The results were validated by immunofluorescence studies. Protein interactions were investigated by yeast two-hybrid assays, coimmunoprecipitation experiments and bimolecular fluorescence complementation. RESULTS: More than 1000 proteins were identified. Among these, 55 proteins were significantly over-represented and 40 proteins were significantly under-represented in targets compared to intraindividual control samples. The majority of over-represented proteins were associated with the myofibrillar Z-disc and actin dynamics, followed by myosin and myosin-associated proteins, proteins involved in protein biosynthesis and chaperones. Under-represented proteins were mainly mitochondrial proteins. Functional studies revealed that the LIM domain of the over-represented protein LIMCH1 interacts with isoform A of Xin actin-binding repeat-containing protein 1 (XinA). CONCLUSIONS: In particular, proteins involved in myofibrillogenesis are over-represented in target structures, which indicate an ongoing process of sarcomere assembly and/or remodelling within this specific area of the muscle fibres. We speculate that target structures are the result of reinnervation processes in which filamin C-associated myofibrillogenesis is tightly regulated by the BAG3-associated protein quality system.


Subject(s)
Peripheral Nervous System Diseases , Humans , Peripheral Nervous System Diseases/metabolism , Actins/analysis , Actins/metabolism , Proteomics , Muscle Proteins/metabolism , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/analysis , Apoptosis Regulatory Proteins/metabolism
3.
Muscle Nerve ; 61(1): 116-121, 2020 01.
Article in English | MEDLINE | ID: mdl-31644823

ABSTRACT

INTRODUCTION: Sporadic inclusion body myositis (sIBM) is characterized by myopathological features including rimmed vacuoles (RVs) and proteins associated with protein aggregation, autophagy, and inflammation. Previous proteomic studies of RV areas revealed an overrepresentation of several chaperones and subunits of the T-complex protein 1 (TCP-1), which is involved in prevention of protein aggregation. METHODS: To validate our proteomic findings, immunofluorescence analyses of selected chaperones and quantitative Western blot analysis of TCP-1 proteins were performed in five sIBM patients and five healthy controls. RESULTS: Immunofluorescence studies confirmed increased immunoreactivity for VCP, UNC45B, GRP-75, αB-crystallin, LAMP-2, Rab-7a, and TCP-1α and TCP-θ in RVs. Quantitative Western blot analysis revealed a significantly higher level of TCP-1 in sIBM muscle tissue when compared with healthy controls. DISCUSSION: Our study findings validate new insights in protein quality control and degradation processes that seem to be relevant in sIBM. These data provide an important basis for future functional and therapeutic studies.


Subject(s)
Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Myositis, Inclusion Body/genetics , Myositis, Inclusion Body/metabolism , Proteomics , Autophagy , Chaperonin Containing TCP-1/genetics , Humans , Inflammation/etiology , Inflammation/pathology , Vacuoles/pathology
4.
Acta Neuropathol Commun ; 7(1): 197, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31796104

ABSTRACT

Protein aggregation is a pathological feature of neurodegenerative disorders. We previously demonstrated that protein inclusions in the brain are composed of supersaturated proteins, which are abundant and aggregation-prone, and form a metastable subproteome. It is not yet clear, however, whether this phenomenon is also associated with non-neuronal protein conformational disorders. To respond to this question, we analyzed proteomic datasets from biopsies of patients with genetic and acquired protein aggregate myopathy (PAM) by quantifying the changes in composition, concentration and aggregation propensity of proteins in the fibers containing inclusions and those surrounding them. We found that a metastable subproteome is present in skeletal muscle from healthy patients. The expression of this subproteome escalate as proteomic samples are taken more proximal to the pathologic inclusion, eventually exceeding its solubility limits and aggregating. While most supersaturated proteins decrease or maintain steady abundance across healthy fibers and inclusion-containing fibers, proteins within the metastable subproteome rise in abundance, suggesting that they escape regulation. Taken together, our results show in the context of a human conformational disorder that the supersaturation of a metastable subproteome underlies widespread aggregation and correlates with the histopathological state of the tissue.


Subject(s)
Inclusion Bodies/metabolism , Myositis, Inclusion Body/metabolism , Proteome/metabolism , Humans , Inclusion Bodies/chemistry , Inclusion Bodies/genetics , Muscular Diseases/metabolism , Muscular Diseases/pathology , Myositis, Inclusion Body/pathology , Protein Aggregates/physiology , Protein Stability , Proteome/analysis , Proteome/genetics
5.
Ann Neurol ; 81(2): 227-239, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28009083

ABSTRACT

OBJECTIVE: Sporadic inclusion body myositis (sIBM) pathogenesis is unknown; however, rimmed vacuoles (RVs) are a constant feature. We propose to identify proteins that accumulate within RVs. METHODS: RVs and intact myofibers were laser microdissected from skeletal muscle of 18 sIBM patients and analyzed by a sensitive mass spectrometry approach using label-free spectral count-based relative protein quantification. Whole exome sequencing was performed on 62 sIBM patients. Immunofluorescence was performed on patient and mouse skeletal muscle. RESULTS: A total of 213 proteins were enriched by >1.5 -fold in RVs compared to controls and included proteins previously reported to accumulate in sIBM tissue or when mutated cause myopathies with RVs. Proteins associated with protein folding and autophagy were the largest group represented. One autophagic adaptor protein not previously identified in sIBM was FYCO1. Rare missense coding FYCO1 variants were present in 11.3% of sIBM patients compared with 2.6% of controls (p = 0.003). FYCO1 colocalized at RVs with autophagic proteins such as MAP1LC3 and SQSTM1 in sIBM and other RV myopathies. One FYCO1 variant protein had reduced colocalization with MAP1LC3 when expressed in mouse muscle. INTERPRETATION: This study used an unbiased proteomic approach to identify RV proteins in sIBM that included a novel protein involved in sIBM pathogenesis. FYCO1 accumulates at RVs, and rare missense variants in FYCO1 are overrepresented in sIBM patients. These FYCO1 variants may impair autophagic function, leading to RV formation in sIBM patient muscle. FYCO1 functionally connects autophagic and endocytic pathways, supporting the hypothesis that impaired endolysosomal degradation underlies the pathogenesis of sIBM. Ann Neurol 2017;81:227-239.


Subject(s)
DNA-Binding Proteins/metabolism , Muscle, Skeletal/metabolism , Myositis, Inclusion Body/metabolism , Proteomics/methods , Transcription Factors/metabolism , Vacuoles/metabolism , Aged , Aged, 80 and over , Alleles , Animals , DNA-Binding Proteins/genetics , Female , Humans , Male , Mice , Microtubule-Associated Proteins , Middle Aged , Myositis, Inclusion Body/genetics , Risk , Transcription Factors/genetics
6.
Neuromuscul Disord ; 27(1): 73-77, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27816332

ABSTRACT

We report three patients with a predominantly proximal myopathy due to p.A193T mutation in the actin-binding domain of FLNC, which has so far only been associated with a distal myopathy. They presented with a late onset myopathy characterized by predominant limb-girdle and proximal weakness. We describe the clinical, electrophysiological, pathological, muscle imaging and genetic features. One of our patients did not have typical histological features for a myofibrillar myopathy in muscle biopsy. This observation is important for the recognition of the full clinical spectrum of filamin-C-related myopathies. Muscle imaging has an important role in distinguishing the different filamin-C myopathy types.


Subject(s)
Filamins/genetics , Muscular Diseases/genetics , Muscular Diseases/physiopathology , Aged , Female , Humans , Middle Aged
7.
Acta Neuropathol ; 132(3): 453-73, 2016 09.
Article in English | MEDLINE | ID: mdl-27393313

ABSTRACT

Secondary mitochondrial dysfunction is a feature in a wide variety of human protein aggregate diseases caused by mutations in different proteins, both in the central nervous system and in striated muscle. The functional relationship between the expression of a mutated protein and mitochondrial dysfunction is largely unknown. In particular, the mechanism how this dysfunction drives the disease process is still elusive. To address this issue for protein aggregate myopathies, we performed a comprehensive, multi-level analysis of mitochondrial pathology in skeletal muscles of human patients with mutations in the intermediate filament protein desmin and in muscles of hetero- and homozygous knock-in mice carrying the R349P desmin mutation. We demonstrate that the expression of mutant desmin causes disruption of the extrasarcomeric desmin cytoskeleton and extensive mitochondrial abnormalities regarding subcellular distribution, number and shape. At the molecular level, we uncovered changes in the abundancy and assembly of the respiratory chain complexes and supercomplexes. In addition, we revealed a marked reduction of mtDNA- and nuclear DNA-encoded mitochondrial proteins in parallel with large-scale deletions in mtDNA and reduced mtDNA copy numbers. Hence, our data demonstrate that the expression of mutant desmin causes multi-level damage of mitochondria already in early stages of desminopathies.


Subject(s)
Desmin/genetics , Intermediate Filaments/pathology , Mitochondria/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Animals , Cytoskeleton/metabolism , Cytoskeleton/pathology , Desmin/metabolism , Humans , Intermediate Filaments/genetics , Mice, Transgenic , Mitochondria/pathology , Muscular Diseases/pathology , Mutation/genetics
8.
Curr Opin Neurol ; 29(5): 628-34, 2016 10.
Article in English | MEDLINE | ID: mdl-27389816

ABSTRACT

PURPOSE OF REVIEW: Myofibrillar myopathies (MFMs) are hereditary muscle disorders characterized by distinct histopathological features. This review provides an overview of recent research with respect to new disease genes, clinical phenotypes, insights into pathomechanisms and therapeutic strategies. RECENT FINDINGS: Beyond the known disease genes DES, FLNC, MYOT, CRYAB, ZASP, BAG3, FHL1 and TTN, mutations in PLEC, ACTA1, HSPB8 and DNAJB6 have also been associated with a MFM phenotype. Proteomic analysis revealed new information about the composition of protein aggregates in myotilinopathy and identified a new diagnostic marker. New animal models mirror central aspects of MFM pathology and novel therapeutic strategies for treatment of MFM were evaluated in cell and animal models. SUMMARY: MFMs are an expanding and numerically significant group of protein aggregate diseases with marked clinical and genetic heterogeneity. Though no specific therapy is currently available, the generation of patient-mimicking cell and animal models now paves the way for the preclinical evaluation of novel therapeutic strategies.


Subject(s)
Muscle, Skeletal/pathology , Mutation , Myopathies, Structural, Congenital/diagnosis , Phenotype , Proteomics , Animals , Disease Models, Animal , Humans , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/metabolism , Myopathies, Structural, Congenital/pathology
9.
Hum Mol Genet ; 24(25): 7207-20, 2015 Dec 20.
Article in English | MEDLINE | ID: mdl-26472074

ABSTRACT

Filamin C (FLNC) mutations in humans cause myofibrillar myopathy (MFM) and cardiomyopathy, characterized by protein aggregation and myofibrillar degeneration. We generated the first patient-mimicking knock-in mouse harbouring the most common disease-causing filamin C mutation (p.W2710X). These heterozygous mice developed muscle weakness and myofibrillar instability, with formation of filamin C- and Xin-positive lesions streaming between Z-discs. These lesions, which are distinct from the classical MFM protein aggregates by their morphology and filamentous appearance, were greatly increased in number upon acute physical exercise in the mice. This pathology suggests that mutant filamin influences the mechanical stability of myofibrillar Z-discs, explaining the muscle weakness in mice and humans. Re-evaluation of biopsies from MFM-filaminopathy patients with different FLNC mutations revealed a similar, previously unreported lesion pathology, in addition to the classical protein aggregates, and suggested that structures previously interpreted as aggregates may be in part sarcomeric lesions. We postulate that these lesions define preclinical disease stages, preceding the formation of protein aggregates.


Subject(s)
Muscle, Skeletal/pathology , Myofibrils/pathology , Animals , Filamins/genetics , Genotype , Mice , Microscopy, Electron , Muscular Diseases/genetics , Muscular Diseases/pathology , Muscular Dystrophies/genetics , Myofibrils/genetics , Phenotype
10.
Neuromuscul Disord ; 25(9): 719-24, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26231297

ABSTRACT

Homozygosity for the common Caucasian splice site mutation c.-32-13T>G in intron 1 of the GAA gene is rather rare in Pompe patients. We report on the clinical, biochemical, morphological, muscle imaging, and genetic findings of six adult Pompe patients from five unrelated families with the c.-32-13T>G GAA gene mutation in homozygous state. All patients had decreased GAA activity and elevated creatine kinase levels. Five patients, aged between 43 and 61 years (median 53 years), initially presented with myalgia, hyperCKaemia, and/or exercise induced fatigue at an age of onset (12-55 years). All but one had proximal lower limb weakness combined with axial weakness and moderate respiratory insufficiency; the sixth patient presented with hyperCKaemia only. Muscle biopsies showed PAS-positive vacuolar myopathy with lysosomal changes and reduced GAA activity. Muscle MRI of lower limb muscles revealed a moderate adipose substitution of the gluteal muscles, biceps femoris and slight fatty infiltration of all thigh muscles. One MRI of the respiratory muscles revealed a diaphragmatic atrophy with unilateral diaphragm elevation. So, the common Caucasian, so called mild, splice site mutation c.-32-13T>G in intron 1 of the GAA gene in a homozygote status reflects the full adult Pompe disease phenotype severity spectrum.


Subject(s)
Glycogen Storage Disease Type II/diagnosis , Glycogen Storage Disease Type II/genetics , alpha-Glucosidases/genetics , Adult , Disease Progression , Female , Glycogen Storage Disease Type II/complications , Homozygote , Humans , Male , Middle Aged , Muscle, Skeletal/pathology , Mutation , Phenotype , RNA Splice Sites
11.
Muscle Nerve ; 52(6): 956-62, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25808807

ABSTRACT

INTRODUCTION: In sporadic inclusion-body myositis (IBM), additional tools are needed to confirm the diagnosis, particularly in clinically atypical or pathologically unproven patients. The aims of this study were to define the pattern of muscle MRI in IBM and to assess its accuracy in differentiating IBM from other myopathies that overlap with it clinically or pathologically. METHODS: Blind assessment was done on the scans of 17 definite IBM, 2 possible IBM, and 118 patients with other myopathies. RESULTS: The diagnostic accuracy to detect definite IBM was 95% for the typical pattern (with 100% specificity) and 97% for both typical and consistent patterns (with 97% specificity). CONCLUSIONS: Muscle MRI is an accurate tool for diagnostic work-up of suspected IBM patients and may be particularly helpful in patients with early disease or who lack the classical IBM pathology.


Subject(s)
Magnetic Resonance Imaging , Muscle, Skeletal/pathology , Myositis, Inclusion Body/diagnosis , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Myositis, Inclusion Body/classification , Myositis, Inclusion Body/physiopathology , Sensitivity and Specificity
12.
Neuromuscul Disord ; 25(5): 392-6, 2015 May.
Article in English | MEDLINE | ID: mdl-25740301

ABSTRACT

Congenital myopathies are clinically and genetically heterogeneous disorders, which often remain genetically undiagnosed for many years. Here we present a 40-year old patient with an almost lifelong history of a congenital myopathy of unknown cause. Muscle biopsy in childhood revealed mild myopathic features and rods. Clinical examination on presentation at the age of 40 revealed a facial weakness, atrophy and weakness of the arm muscles and distal leg muscles with mild contractures of the foot flexors and the right elbow. Subsequently, the nebulin gene was identified as a putative candidate gene by linkage analyses, but sequence analysis only revealed one heterozygous splice site mutation in intron 73 (c.10872+1G>T). Therefore, "Next Generation Sequencing" was performed, which revealed a second pathogenic variant in exon 145 (c.21622A>C). Compound-heterozygous carrier status was confirmed via sequence analysis of the index patient's parents. Whole body muscle MRI showed a muscle involvement as previously described in nebulin-associated myopathies. Based on biopsy material, genetic analyses and muscle MRI, we identified two novel, compound-heterozygous variants in the nebulin gene after a 30 year clinical history, which cause a classical childhood type of nemaline myopathy.


Subject(s)
Muscle Proteins/genetics , Muscle, Skeletal/pathology , Myopathies, Nemaline/genetics , Adult , Humans , Magnetic Resonance Imaging , Male , Muscle, Skeletal/metabolism , Mutation
13.
Neuropathol Appl Neurobiol ; 41(7): 952-63, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25711697

ABSTRACT

OBJECTIVE: To analyse the paradox of a lack of giant cell formation and fibrosis in chronic lesions of macrophagic myofasciitis (MMF) in comparison with muscular sarcoidosis (MuS). METHODS: Inflammatory lesions and contiguous muscle regions from biopsy samples of 10 patients with MuS and 10 patients with MMF were cut out by laser microdissection. Mediators of the T helper cell (Th)1 inducing classical macrophage activation (e.g. STAT1, IFNγ and CXCR3), and Th2 inducing alternative activation of macrophages (e.g. CD206/MRC1, STAT6, SOCS1), molecules involved in development of fibrosis (e.g. TGFß) and giant cells (e.g. TYROBP), were assessed by immunohistochemistry and real-time polymerase chain reaction (PCR). RESULTS: STAT6-induced Th2 immunity was associated with up-regulated gene expression of MRC1, SOCS1 and TGFB in inflammatory foci, in comparison with adjacent tissue. TYROBP and TREM2, genes regulating giant cell formation, were more strongly expressed in lesions of MuS patients than in those of MMF. TGFß co-localized with CD206(+) macrophages in MuS but not in MMF. Conversely, Th1 immunity was illustrated by STAT1 staining both in macrophages and myofibres in MuS, but not in MMF. Also, STAT1-induced IFNG and CXCR3 expression in lesions and the surrounding tissue was elevated compared with normal controls, but without statistically significant differences. CONCLUSION: Giant cell and typical granuloma formations, including fibrogenesis, is dependent on two main mechanisms, both involving specific macrophage activation: a strong Th2-M2 polarization and a significant expression of TYROBP and TGFß in macrophages. The low-grade alternative activation of macrophages in MMF lesions and poor TYROBP and TGFßco-expression are obviously insufficient to produce giant cells.


Subject(s)
Fasciitis/immunology , Macrophages/immunology , Muscle, Skeletal/immunology , Myositis/immunology , Sarcoidosis/immunology , Th2 Cells/immunology , Adult , Aged , Fasciitis/pathology , Female , Humans , Macrophage Activation , Macrophages/pathology , Male , Middle Aged , Muscle, Skeletal/pathology , Myositis/pathology , Sarcoidosis/pathology , Th2 Cells/pathology , Young Adult
14.
Orphanet J Rare Dis ; 9: 121, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25208129

ABSTRACT

BACKGROUND: Myofibrillar myopathies (MFM) are a group of phenotypically and genetically heterogeneous neuromuscular disorders, which are characterized by protein aggregations in muscle fibres and can be associated with multisystemic involvement. METHODS: We screened a large cohort of 38 index patients with MFM for mutations in the nine thus far known causative genes using Sanger and next generation sequencing (NGS). We studied the clinical and histopathological characteristics in 38 index patients and five additional relatives (n = 43) and particularly focused on the associated multisystemic symptoms. RESULTS: We identified 14 heterozygous mutations (diagnostic yield of 37%), among them the novel p.Pro209Gln mutation in the BAG3 gene, which was associated with onset in adulthood, a mild phenotype and an axonal sensorimotor polyneuropathy, in the absence of giant axons at the nerve biopsy. We revealed several novel clinical phenotypes and unusual multisystemic presentations with previously described mutations: hearing impairment with a FLNC mutation, dysphonia with a mutation in DES and the first patient with a FLNC mutation presenting respiratory insufficiency as the initial symptom. Moreover, we described for the first time respiratory insufficiency occurring in a patient with the p.Gly154Ser mutation in CRYAB. Interestingly, we detected a polyneuropathy in 28% of the MFM patients, including a BAG3 and a MYOT case, and hearing impairment in 13%, including one patient with a FLNC mutation and two with mutations in the DES gene. In four index patients with a mutation in one of the MFM genes, typical histological findings were only identified at the ultrastructural level (29%). CONCLUSIONS: We conclude that extraskeletal symptoms frequently occur in MFM, particularly cardiac and respiratory involvement, polyneuropathy and/or deafness. BAG3 mutations should be considered even in cases with a mild phenotype or an adult onset. We identified a genetic defect in one of the known genes in less than half of the MFM patients, indicating that more causative genes are still to be found. Next generation sequencing techniques should be helpful in achieving this aim.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , High-Throughput Nucleotide Sequencing/methods , Muscle, Skeletal/metabolism , Mutation , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Muscle, Skeletal/pathology , Myopathies, Structural, Congenital/complications , Myopathies, Structural, Congenital/genetics , Pedigree , Phenotype , Young Adult
15.
JIMD Rep ; 17: 53-61, 2014.
Article in English | MEDLINE | ID: mdl-25155777

ABSTRACT

OBJECTIVE: To determine the frequency and impact of gastrointestinal symptoms, and bowel and urinary incontinence, as this is currently unknown in adults with Pompe disease. METHODS: Adult German Pompe patients and age- and gender-matched controls were asked about symptoms in the upper and lower intestinal tract as well as urinary incontinence using the Gastrointestinal Symptoms Questionnaire and the International Consultation on Incontinence Questionnaires for Bowel Symptoms and Urinary Incontinence. RESULTS: The overall response rate was 78%; 57 patients and 57 controls participated. The mean age of the patients was 48.3 years ±14.7 (28 female, 29 male). 84% of patients were receiving enzyme replacement therapy. Stool urgency, diarrhoea, and urinary urge incontinence were reported significantly more frequently in patients compared to the age- and gender-matched controls (55%, 56%, 33% vs. 20%, 18%, 7%). 20% of Pompe patients used loperamide daily against diarrhoea. No other gastrointestinal tract-related symptoms were reported to occur more frequently in Pompe patients than in controls. CONCLUSIONS: Compared to age- and gender-matched controls, both urinary and bowel incontinence occur in a higher frequency in adults with Pompe disease and have a major impact on daily life.

16.
Mol Genet Metab ; 112(2): 148-53, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24726296

ABSTRACT

BACKGROUND: The obstetric risk in patients with Pompe disease (glycogen storage disease type II), a mainly skeletal muscle disorder, is unknown. METHODS: The clinical course and the outcome of pregnancy, and the effect of pregnancy on disease manifestations or clinical signs and symptoms in Pompe disease were analyzed retrospectively using a questionnaire. Participating women with Pompe disease were recruited by the German and the UK sections of the International Pompe Association, and by centers associated within the German Pompe Group. The data was compared with information from the German statistical almanac, perinatal registry, and perinatal quality survey. RESULTS: 66 of 136 women responded to the questionnaire (median age: 47 years, range: 18-74). In 10 of 52 women who had been pregnant, the symptoms of Pompe disease were present during pregnancy (n=7 1st, n=1 2nd, n=1 3rd pregnancy). Muscle weakness worsened in 3 women, and first presented in 3 others during the first pregnancy (4.5% each). Respiratory problems deteriorated in 2/10 women during pregnancy. These 10 symptomatic women had 17 pregnancies (15 deliveries, 2 miscarriages, no abortions). The 42 asymptomatic women (63.6%) had 109 pregnancies (72.4% deliveries, 19.3% miscarriages, 7.3% abortions). There were no significant differences between the mean duration of pregnancies or the mean birth weight in symptomatic and asymptomatic women, or compared to the data from the general population. The same was true of pregnancy and delivery complications (including Cesarean section). CONCLUSIONS: Our data show that women with Pompe disease do not appear to have an increased risk of pregnancy or delivery complications. However, muscle weakness and respiratory complications might manifest or worsen during pregnancy in some women.


Subject(s)
Glycogen Storage Disease Type II/complications , Glycogen Storage Disease Type II/epidemiology , Obstetric Labor Complications/epidemiology , Pregnancy Complications/epidemiology , Pregnancy Outcome/epidemiology , Adolescent , Adult , Aged , Delivery, Obstetric/statistics & numerical data , Female , Germany , Glycogen Storage Disease Type II/pathology , Humans , Middle Aged , Pregnancy , Pregnancy Complications/physiopathology , Retrospective Studies , Surveys and Questionnaires , United Kingdom , Young Adult
17.
Innate Immun ; 20(1): 49-60, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23608825

ABSTRACT

Sporadic inclusion body myositis (sIBM) and polymyositis (PM) are characterized by muscle inflammation, with sIBM showing additional degenerative alterations. In this study we investigated human beta defensins and associated TLRs to elucidate the role of the innate immune system in idiopathic inflammatory myopathies (IIM), and its association with inflammatory and degenerative alterations. Expression levels of human beta-defensin (HBD)-1, HBD-2, HBD-3 and TLR2, 3, 4, 7 and 9 were analysed by quantitative real-time PCR in skeletal muscle tissue. Localization of HBD-3, collagen 6, dystrophin, CD8-positive T-cells, CD-68-positive macrophages, ß-amyloid, the autophagy marker LC3, and TLR3 were detected by immunofluorescence and co-localization was quantified. HBD-3 and all TLRs except for TLR9 were overexpressed in both IIM with significant overexpression of TLR3 in sIBM. HBD-3 showed characteristic intracellular accumulations near deposits of ß-amyloid, LC3 and TLR3 in sIBM, and was detected in inflammatory infiltrations and macrophages invading necrotic muscle fibres in both IIM. The characteristic intracellular localization of HBD-3 near markers of degeneration and autophagy, and overexpression of endosomal TLR3 in sIBM hint at different pathogenetic mechanisms in sIBM compared with PM. This descriptive study serves as a first approach to the role of the innate immune system in sIBM and PM.


Subject(s)
Amyloid beta-Peptides/metabolism , Endosomes/metabolism , Microtubule-Associated Proteins/metabolism , Muscle Fibers, Skeletal/pathology , Myositis/immunology , Toll-Like Receptor 3/metabolism , beta-Defensins/metabolism , Adolescent , Adult , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Autophagy , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Collagen Type VI/metabolism , Dystrophin/metabolism , Female , Humans , Immunity, Innate , Macrophages/immunology , Male , Middle Aged , Muscle Fibers, Skeletal/immunology , Myositis/physiopathology , Necrosis , Protein Transport , Toll-Like Receptor 3/genetics , Young Adult , beta-Defensins/genetics
18.
Am J Pathol ; 183(6): 1703-1709, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24225086

ABSTRACT

Xin is a striated muscle-specific protein that is localized to the myotendinous junction in skeletal muscle. However, in injured mouse muscle, Xin expression is up-regulated and observed throughout skeletal muscle fibers and within satellite cells. In this study, Xin was analyzed by immunofluorescent staining in skeletal muscle samples from 47 subjects with various forms of myopathy, including muscular dystrophies, inflammatory myopathies, mitochondrial/metabolic myopathy, and endocrine myopathy. Results indicate that Xin immunoreactivity is positively and significantly correlated (rs = 0.6175, P = <0.0001) with the severity of muscle damage, regardless of myopathy type. Other muscle damage measures also showed a correlation with severity [Xin actin-binding repeat-containing 2 (rs = -0.7108, P = 0.0006) and collagen (rs = 0.4683, P = 0.0783)]. However, because only Xin lacked immunoreactivity within the healthy muscle belly, any detectable immunoreactivity for Xin was indicative of muscle damage. We also investigated the expression of Xin within the skeletal muscle of healthy individuals subjected to damaging eccentric exercise. Consistent with our previously mentioned results, Xin immunoreactivity was increased 24 hours after exercise in damaged muscle fibers and within the activated muscle satellite cells. Taken together, these data demonstrate Xin as a useful biomarker of muscle damage in healthy individuals and in patients with myopathy. The strong correlation between the degree of muscle damage and Xin immunoreactivity suggests that Xin may be a suitable outcome measure to evaluate disease progression and treatment effects in clinical trials.


Subject(s)
DNA-Binding Proteins/metabolism , Muscle, Skeletal , Muscular Diseases/metabolism , Muscular Diseases/pathology , Nuclear Proteins/metabolism , Adult , Aged , Aged, 80 and over , Animals , Biomarkers/metabolism , DNA-Binding Proteins/genetics , Female , Humans , Male , Mice , Middle Aged , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Nuclear Proteins/genetics , Severity of Illness Index
19.
Curr Opin Neurol ; 26(5): 527-35, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23995273

ABSTRACT

PURPOSE OF REVIEW: Myofibrillar myopathies (MFMs) are a heterogeneous group of skeletal and cardiac muscle diseases. In this review, we highlight recent discoveries of new genes and disease mechanisms involved in this group of disorders. RECENT FINDINGS: The advent of next-generation sequencing technology, laser microdissection and mass spectrometry-based proteomics has facilitated the discovery of new MFM causative genes and pathomechanisms. New mutations have also been discovered in 'older' genes, helping to find a classification niche for MFM-linked disorders showing variant phenotypes. Cell transfection experiments using primary cultured myoblasts and newer animal models provide insights into the pathogenesis of MFMs. SUMMARY: An increasing number of genes are involved in the causation of variant subtypes of MFM. The application of modern technologies in combination with classical histopathological and ultrastructural studies is helping to establish the molecular diagnosis and reach a better understanding of the pathogenic mechanisms of each MFM subtype, thus putting an emphasis on the development of specific means for prevention and therapy of these incapacitating and frequently fatal diseases.


Subject(s)
Muscle, Skeletal/pathology , Animals , Genetic Predisposition to Disease/genetics , Humans , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Mutation/genetics , Myopathies, Structural, Congenital/diagnosis , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/therapy , Phenotype
20.
Mol Biol Cell ; 24(20): 3215-26, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23985323

ABSTRACT

The Xin actin-binding repeat-containing proteins Xin and XIRP2 are exclusively expressed in striated muscle cells, where they are believed to play an important role in development. In adult muscle, both proteins are concentrated at attachment sites of myofibrils to the membrane. In contrast, during development they are localized to immature myofibrils together with their binding partner, filamin C, indicating an involvement of both proteins in myofibril assembly. We identify the SH3 domains of nebulin and nebulette as novel ligands of proline-rich regions of Xin and XIRP2. Precise binding motifs are mapped and shown to bind both SH3 domains with micromolar affinity. Cocrystallization of the nebulette SH3 domain with the interacting XIRP2 peptide PPPTLPKPKLPKH reveals selective interactions that conform to class II SH3 domain-binding peptides. Bimolecular fluorescence complementation experiments in cultured muscle cells indicate a temporally restricted interaction of Xin-repeat proteins with nebulin/nebulette during early stages of myofibril development that is lost upon further maturation. In mature myofibrils, this interaction is limited to longitudinally oriented structures associated with myofibril development and remodeling. These data provide new insights into the role of Xin actin-binding repeat-containing proteins (together with their interaction partners) in myofibril assembly and after muscle damage.


Subject(s)
Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Myofibrils/metabolism , Nuclear Proteins/metabolism , Animals , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Humans , LIM Domain Proteins/chemistry , LIM Domain Proteins/genetics , Ligands , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Myofibrils/chemistry , Myofibrils/ultrastructure , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , src Homology Domains/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...