Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Cell Mol Life Sci ; 79(1): 1, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34910257

ABSTRACT

Intestinal mesenchymal cells encompass multiple subsets, whose origins, functions, and pathophysiological importance are still not clear. Here, we used the Col6a1Cre mouse, which targets distinct fibroblast subsets and perivascular cells that can be further distinguished by the combination of the CD201, PDGFRα and αSMA markers. Developmental studies revealed that the Col6a1Cre mouse also targets mesenchymal aggregates that are crucial for intestinal morphogenesis and patterning, suggesting an ontogenic relationship between them and homeostatic PDGFRαhi telocytes. Cell depletion experiments in adulthood showed that Col6a1+/CD201+ mesenchymal cells regulate homeostatic enteroendocrine cell differentiation and epithelial proliferation. During acute colitis, they expressed an inflammatory and extracellular matrix remodelling gene signature, but they also retained their properties and topology. Notably, both in homeostasis and tissue regeneration, they were dispensable for normal organ architecture, while CD34+ mesenchymal cells expanded, localised at the top of the crypts, and showed increased expression of villous-associated morphogenetic factors, providing thus evidence for the plasticity potential of intestinal mesenchymal cells. Our results provide a comprehensive analysis of the identities, origin, and functional significance of distinct mesenchymal populations in the intestine.


Subject(s)
Collagen Type VI/metabolism , Endothelial Protein C Receptor/metabolism , Intestines/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Plasticity , Cell Proliferation , Colitis/chemically induced , Colitis/pathology , Collagen Type VI/deficiency , Collagen Type VI/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Intestines/cytology , Intestines/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Regeneration
3.
Neuromuscul Disord ; 27(3): 278-285, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28087121

ABSTRACT

Congenital muscular dystrophy (CMD) comprises a rare group of genetic muscle diseases that present at birth or early during infancy. Two common subtypes of CMD are collagen VI-related muscular dystrophy (COL6-RD) and laminin alpha 2-related dystrophy (LAMA2-RD). Traditional outcome measures in CMD include gross motor and mobility assessments, yet significant motor declines underscore the need for valid upper extremity motor assessments as a clinical endpoint. This study validated a battery of upper extremity measures in these two CMD subtypes for future clinical trials. For this cross-sectional study, 42 participants were assessed over the same 2-5 day period at the National Institutes of Health Clinical Center. All upper extremity measures were correlated with the Motor Function Measure 32 (MFM32). The battery of upper extremity assessments included the Jebsen Taylor Hand Function Test, Quality of Upper Extremity Skills Test (QUEST), hand held dynamometry, goniometry, and MyoSet Tools. Spearman Rho was used for correlations to the MFM32. Pearson was performed to correlate the Jebsen, QUEST, hand-held dynamometry, goniometry and the MyoSet Tools. Correlations were considered significant at the 0.01 level (2-tailed). Significant correlations were found between both the MFM32 and MFM Dimension 3 only (Distal Motor function) and the Jebsen, QUEST, MyoGrip and MyoPinch, elbow flexion/extension ROM and myometry. Additional correlations between the assessments are reported. The Jebsen, the Grasp and Dissociated Movements domains of the QUEST, the MyoGrip and the MyoPinch tools, as well as elbow ROM and myometry were determined to be valid and feasible in this population, provided variation in test items, and assessed a range of difficulty in CMD. To move forward, it will be of utmost importance to determine whether these upper extremity measures are reproducible and sensitive to change over time.


Subject(s)
Collagen Type VI , Laminin , Muscular Dystrophies/diagnosis , Severity of Illness Index , Upper Extremity/physiopathology , Adolescent , Adult , Child , Child, Preschool , Collagen Type VI/deficiency , Cross-Sectional Studies , Female , Humans , Laminin/deficiency , Male , Muscular Dystrophies/congenital , Young Adult
4.
EBioMedicine ; 15: 193-202, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28043812

ABSTRACT

Congenital muscular dystrophies with collagen VI deficiency are inherited muscle disorders with a broad spectrum of clinical presentation and are caused by mutations in one of COL6A1-3 genes. Muscle pathology is characterized by fiber size variation and increased interstitial fibrosis and adipogenesis. In this study, we define critical events that contribute to muscle weakness and fibrosis in a mouse model with collagen VI deficiency. The Col6a1GT/GT mice develop non-progressive weakness from younger age, accompanied by stunted muscle growth due to reduced IGF-1 signaling activity. In addition, the Col6a1GT/GT mice have high numbers of interstitial skeletal muscle mesenchymal progenitor cells, which dramatically increase with repeated myofiber necrosis/regeneration. Our results suggest that impaired neonatal muscle growth and the activation of the mesenchymal cells in skeletal muscles contribute to the pathology of collagen VI deficient muscular dystrophy, and more importantly, provide the insights on the therapeutic strategies for collagen VI deficiency.


Subject(s)
Collagen Type VI/deficiency , Muscle Weakness/genetics , Muscle Weakness/pathology , Muscular Dystrophies/genetics , Muscular Dystrophies/physiopathology , Alleles , Animals , Disease Models, Animal , Disease Progression , Fibrosis , Genotype , Humans , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Transgenic , Muscle Contraction/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophies/pathology , Muscular Dystrophy, Animal , Mutation , Phenotype , Signal Transduction
5.
Ann Neurol ; 80(1): 101-11, 2016 07.
Article in English | MEDLINE | ID: mdl-27159402

ABSTRACT

OBJECTIVE: To evaluate the diagnostic outcomes in a large cohort of congenital muscular dystrophy (CMD) patients using traditional and next generation sequencing (NGS) technologies. METHODS: A total of 123 CMD patients were investigated using the traditional approaches of histology, immunohistochemical analysis of muscle biopsy, and candidate gene sequencing. Undiagnosed patients available for further testing were investigated using NGS. RESULTS: Muscle biopsy and immunohistochemical analysis found deficiencies of laminin α2, α-dystroglycan, or collagen VI in 50% of patients. Candidate gene sequencing and chromosomal microarray established a genetic diagnosis in 32% (39 of 123). Of 85 patients presenting in the past 20 years, 28 of 51 who lacked a confirmed genetic diagnosis (55%) consented to NGS studies, leading to confirmed diagnoses in a further 11 patients. Using the combination of approaches, a confirmed genetic diagnosis was achieved in 51% (43 of 85). The diagnoses within the cohort were heterogeneous. Forty-five of 59 probands with confirmed or probable diagnoses had variants in genes known to cause CMD (76%), and 11 of 59 (19%) had variants in genes associated with congenital myopathies, reflecting overlapping features of these conditions. One patient had a congenital myasthenic syndrome, and 2 had microdeletions. Within the cohort, 5 patients had variants in novel (PIGY and GMPPB) or recently published genes (GFPT1 and MICU1), and 7 had variants in TTN or RYR1, large genes that are technically difficult to Sanger sequence. INTERPRETATION: These data support NGS as a first-line tool for genetic evaluation of patients with a clinical phenotype suggestive of CMD, with muscle biopsy reserved as a second-tier investigation. Ann Neurol 2016;80:101-111.


Subject(s)
Genetic Predisposition to Disease/genetics , Muscular Dystrophies/diagnosis , Muscular Dystrophies/genetics , Adolescent , Adult , Child , Child, Preschool , Collagen Type VI/deficiency , Dystroglycans/deficiency , Genetic Variation/genetics , High-Throughput Nucleotide Sequencing , Humans , Infant , Laminin/deficiency , Muscle, Skeletal/metabolism , Young Adult
6.
Autophagy ; 11(12): 2142-52, 2015.
Article in English | MEDLINE | ID: mdl-26565691

ABSTRACT

Autophagy is a self-degradative process responsible for the clearance of damaged or unnecessary cellular components. We have previously found that persistence of dysfunctional organelles due to autophagy failure is a key event in the pathogenesis of COL6/collagen VI-related myopathies, and have demonstrated that reactivation of a proper autophagic flux rescues the muscle defects of Col6a1-null (col6a1(-/-)) mice. Here we show that treatment with spermidine, a naturally occurring nontoxic autophagy inducer, is beneficial for col6a1(-/-) mice. Systemic administration of spermidine in col6a1(-/-) mice reactivated autophagy in a dose-dependent manner, leading to a concurrent amelioration of the histological and ultrastructural muscle defects. The beneficial effects of spermidine, together with its being easy to administer and the lack of overt side effects, open the field for the design of novel nutraceutical strategies for the treatment of muscle diseases characterized by autophagy impairment.


Subject(s)
Autophagy/genetics , Collagen Type VI/deficiency , Muscles/metabolism , Muscular Diseases/pathology , Spermidine/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Autophagy/physiology , Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/pathology , Muscles/pathology
8.
PLoS One ; 8(10): e77430, 2013.
Article in English | MEDLINE | ID: mdl-24223098

ABSTRACT

Ullrich congenital muscular dystrophy (UCMD), caused by collagen VI deficiency, is a common congenital muscular dystrophy. At present, the role of collagen VI in muscle and the mechanism of disease are not fully understood. To address this we have applied microarrays to analyse the transcriptome of UCMD muscle and compare it to healthy muscle and other muscular dystrophies. We identified 389 genes which are differentially regulated in UCMD relative to controls. In addition, there were 718 genes differentially expressed between UCMD and dystrophin deficient muscle. In contrast, only 29 genes were altered relative to other congenital muscular dystrophies. Changes in gene expression were confirmed by real-time PCR. The set of regulated genes was analysed by Gene Ontology, KEGG pathways and Ingenuity Pathway analysis to reveal the molecular functions and gene networks associated with collagen VI defects. The most significantly regulated pathways were those involved in muscle regeneration, extracellular matrix remodelling and inflammation. We characterised the immune response in UCMD biopsies as being mainly mediated via M2 macrophages and the complement pathway indicating that anti-inflammatory treatment may be beneficial to UCMD as for other dystrophies. We studied the immunolocalisation of ECM components and found that biglycan, a collagen VI interacting proteoglycan, was reduced in the basal lamina of UCMD patients. We propose that biglycan reduction is secondary to collagen VI loss and that it may be contributing towards UCMD pathophysiology. Consequently, strategies aimed at over-expressing biglycan and restore the link between the muscle cell surface and the extracellular matrix should be considered.


Subject(s)
Collagen Type VI/deficiency , Muscular Dystrophies/metabolism , Sclerosis/metabolism , Transcriptome , Case-Control Studies , Child , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Extracellular Matrix/metabolism , Gene Expression Profiling , Genes, MHC Class II , Humans , Mitochondria/genetics , Mitochondria/metabolism , Molecular Sequence Annotation , Molecular Targeted Therapy , Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , Oligonucleotide Array Sequence Analysis , Proteolysis , Sclerosis/genetics , Sclerosis/therapy
9.
Brain ; 136(Pt 12): 3625-33, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24271325

ABSTRACT

The spectrum of clinical phenotypes associated with a deficiency or dysfunction of collagen VI in the extracellular matrix of muscle are collectively termed 'collagen VI-related myopathies' and include Ullrich congenital muscular dystrophy, Bethlem myopathy and intermediate phenotypes. To further define the clinical course of these variants, we studied the natural history of pulmonary function in correlation to motor abilities in the collagen VI-related myopathies by analysing longitudinal forced vital capacity data in a large international cohort. Retrospective chart reviews of genetically and/or pathologically confirmed collagen VI-related myopathy patients were performed at 10 neuromuscular centres: USA (n = 2), UK (n = 2), Australia (n = 2), Italy (n = 2), France (n = 1) and Belgium (n = 1). A total of 486 forced vital capacity measurements obtained in 145 patients were available for analysis. Patients at the severe end of the clinical spectrum, conforming to the original description of Ullrich congenital muscular dystrophy were easily identified by severe muscle weakness either preventing ambulation or resulting in an early loss of ambulation, and demonstrated a cumulative decline in forced vital capacity of 2.6% per year (P < 0.0001). Patients with better functional abilities, in whom walking with/without assistance was achieved, were initially combined, containing both intermediate and Bethlem myopathy phenotypes in one group. However, one subset of patients demonstrated a continuous decline in pulmonary function whereas the other had stable pulmonary function. None of the patients with declining pulmonary function attained the ability to hop or run; these patients were categorized as intermediate collagen VI-related myopathy and the remaining patients as Bethlem myopathy. Intermediate patients had a cumulative decline in forced vital capacity of 2.3% per year (P < 0.0001) whereas the relationship between age and forced vital capacity in patients with Bethlem myopathy was not significant (P = 0.1432). Nocturnal non-invasive ventilation was initiated in patients with Ullrich congenital muscular dystrophy by 11.3 years (±4.0) and in patients with intermediate collagen VI-related myopathy by 20.7 years (±1.5). The relationship between maximal motor ability and forced vital capacity was highly significant (P < 0.0001). This study demonstrates that pulmonary function profiles can be used in combination with motor function profiles to stratify collagen VI-related myopathy patients phenotypically. These findings improve our knowledge of the natural history of the collagen VI-related myopathies, enabling proactive optimization of care and preparing this patient population for clinical trials.


Subject(s)
Collagen Type VI/genetics , Lung Diseases/etiology , Muscular Diseases/complications , Muscular Diseases/genetics , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Collagen Type VI/deficiency , Disability Evaluation , Europe , Female , Humans , Kaplan-Meier Estimate , Linear Models , Lung Diseases/genetics , Lung Diseases/therapy , Male , Middle Aged , Motor Activity , Muscular Diseases/classification , Muscular Diseases/epidemiology , Respiration, Artificial , Retrospective Studies , United States , Vital Capacity/genetics , Young Adult
10.
Rev. neurol. (Ed. impr.) ; 57(supl.1): s47-s52, 6 sept., 2013.
Article in Spanish | IBECS | ID: ibc-149005

ABSTRACT

Las distrofias musculares congénitas (DMC) representan desde el punto de vista clínico y genético un grupo heterogéneo de enfermedades dentro de la patología neuromuscular. Las formas más conocidas son: DMC por déficit de merosina, DMC por déficit de colágeno VI, DMC relacionada con LMNA, DMC relacionada con selenoproteína (SEPN1) y las DMC vinculadas a los alfa-distroglicanos. Se presentan con un amplio espectro de fenotipos clínicos. En su mayoría son de herencia autosómica recesiva. Con mucha frecuencia las manifestaciones iniciales comienzan en la infancia o en el período neonatal. Se sospechan clínicamente por la existencia de hipotonía y paresia y se caracterizan por la existencia de un patrón distrófico en la biopsia muscular (sustitución de músculo por tejido fibroadiposo, con necrosis y regeneración celular). Avances en la comprensión de la patogénesis molecular de las DMC han permitido profundizar en la clasificación de los diferentes subtipos. El objetivo de esta revisión es comentar los avances de los últimos años en cuanto a la clasificación de las DMC en relación a la genética, las proteínas involucradas y su presentación clínica (AU)


From the clinical and genetic point of view, congenital muscular dystrophies (CMD) are a heterogenic group of diseases within neuromuscular pathologies. The best known forms are: merosin deficiency CMD, collagen VI deficiency CMD, LMNA-related CMD, selenoprotein-related CMD (SEPN1) and alpha-dystroglycan-related CMD. They present with a broad spectrum of clinical phenotypes. Most of them are transmitted by recessive autosomal inheritance. The initial manifestations very often begin in infancy or in the neonatal period. There are clinical suspicions of the existence of hypotonia and paresis, and they are characterised by a dystrophic pattern in the muscular biopsy (muscle replaced by fibroadipose tissue, with necrosis and cell regeneration). Advances in the understanding of the molecular pathogenesis of CMD have made it possible to make further progress in the classification of the different subtypes. The aim of this review is to comment on the advances made in recent years as regards the classification of CMD in terms of genetics, the proteins involved and their clinical presentation (AU)


Subject(s)
Humans , Child , Muscular Dystrophies/classification , Muscular Dystrophies/congenital , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , Collagen Type VI/deficiency , Collagen Type VI/genetics , Selenoproteins/genetics , Selenoproteins/deficiency , Muscle Proteins/genetics , Muscle Proteins/deficiency , Genotype , Laminin/deficiency , Laminin/genetics , Lamin Type A/deficiency , Lamin Type A/genetics , Dystroglycans/deficiency , Dystroglycans/genetics
11.
Rev Neurol ; 57 Suppl 1: S47-52, 2013 Sep 06.
Article in Spanish | MEDLINE | ID: mdl-23897156

ABSTRACT

From the clinical and genetic point of view, congenital muscular dystrophies (CMD) are a heterogenic group of diseases within neuromuscular pathologies. The best known forms are: merosin deficiency CMD, collagen VI deficiency CMD, LMNA-related CMD, selenoprotein-related CMD (SEPN1) and alpha-dystroglycan-related CMD. They present with a broad spectrum of clinical phenotypes. Most of them are transmitted by recessive autosomal inheritance. The initial manifestations very often begin in infancy or in the neonatal period. There are clinical suspicions of the existence of hypotonia and paresis, and they are characterised by a dystrophic pattern in the muscular biopsy (muscle replaced by fibroadipose tissue, with necrosis and cell regeneration). Advances in the understanding of the molecular pathogenesis of CMD have made it possible to make further progress in the classification of the different subtypes. The aim of this review is to comment on the advances made in recent years as regards the classification of CMD in terms of genetics, the proteins involved and their clinical presentation.


TITLE: Distrofias musculares congenitas en el niño.Las distrofias musculares congenitas (DMC) representan desde el punto de vista clinico y genetico un grupo heterogeneo de enfermedades dentro de la patologia neuromuscular. Las formas mas conocidas son: DMC por deficit de merosina, DMC por deficit de colageno VI, DMC relacionada con LMNA, DMC relacionada con selenoproteina (SEPN1) y las DMC vinculadas a los alfa-distroglicanos. Se presentan con un amplio espectro de fenotipos clinicos. En su mayoria son de herencia autosomica recesiva. Con mucha frecuencia las manifestaciones iniciales comienzan en la infancia o en el periodo neonatal. Se sospechan clinicamente por la existencia de hipotonia y paresia y se caracterizan por la existencia de un patron distrofico en la biopsia muscular (sustitucion de musculo por tejido fibroadiposo, con necrosis y regeneracion celular). Avances en la comprension de la patogenesis molecular de las DMC han permitido profundizar en la clasificacion de los diferentes subtipos. El objetivo de esta revision es comentar los avances de los ultimos años en cuanto a la clasificacion de las DMC en relacion a la genetica, las proteinas involucradas y su presentacion clinica.


Subject(s)
Muscular Dystrophies/congenital , Child , Collagen Type VI/deficiency , Collagen Type VI/genetics , Dystroglycans/deficiency , Dystroglycans/genetics , Genotype , Humans , Lamin Type A/deficiency , Lamin Type A/genetics , Laminin/deficiency , Laminin/genetics , Muscle Proteins/deficiency , Muscle Proteins/genetics , Muscular Dystrophies/classification , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , Selenoproteins/deficiency , Selenoproteins/genetics
12.
Nat Commun ; 4: 1964, 2013.
Article in English | MEDLINE | ID: mdl-23743995

ABSTRACT

Adult muscle stem cells, or satellite cells have essential roles in homeostasis and regeneration of skeletal muscles. Satellite cells are located within a niche that includes myofibers and extracellular matrix. The function of specific extracellular matrix molecules in regulating SCs is poorly understood. Here, we show that the extracellular matrix protein collagen VI is a key component of the satellite cell niche. Lack of collagen VI in Col6a1(-/-) mice causes impaired muscle regeneration and reduced satellite cell self-renewal capability after injury. Collagen VI null muscles display significant decrease of stiffness, which is able to compromise the in vitro and in vivo activity of wild-type satellite cells. When collagen VI is reinstated in vivo by grafting wild-type fibroblasts, the biomechanical properties of Col6a1(-/-) muscles are ameliorated and satellite cell defects rescued. Our findings establish a critical role for an extracellular matrix molecule in satellite cell self-renewal and open new venues for therapies of collagen VI-related muscle diseases.


Subject(s)
Collagen Type VI/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/cytology , Animals , Cell Proliferation , Collagen Type VI/deficiency , Elastic Modulus , Extracellular Space/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/transplantation , Fluorescent Antibody Technique , In Vitro Techniques , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Satellite Cells, Skeletal Muscle/metabolism , Stem Cell Niche , Transcription Factors/metabolism
13.
J Biol Chem ; 288(20): 14320-14331, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23564457

ABSTRACT

Collagen VI is a ubiquitously expressed extracellular microfibrillar protein. Its most common molecular form is composed of the α1(VI), α2(VI), and α3(VI) collagen α chains encoded by the COL6A1, COL6A2, and COL6A3 genes, respectively. Mutations in any of the three collagen VI genes cause congenital muscular dystrophy types Bethlem and Ullrich as well as intermediate phenotypes characterized by muscle weakness and connective tissue abnormalities. The α3(VI) collagen α chain has much larger N- and C-globular domains than the other two chains. Its most C-terminal domain can be cleaved off after assembly into microfibrils, and the cleavage product has been implicated in tumor angiogenesis and progression. Here we characterize a Col6a3 mutant mouse that expresses a very low level of a non-functional α3(VI) collagen chain. The mutant mice are deficient in extracellular collagen VI microfibrils and exhibit myopathic features, including decreased muscle mass and contractile force. Ultrastructurally abnormal collagen fibrils were observed in tendon, but not cornea, of the mutant mice, indicating a distinct tissue-specific effect of collagen VI on collagen I fibrillogenesis. Overall, the mice lacking normal α3(VI) collagen chains displayed mild musculoskeletal phenotypes similar to mice deficient in the α1(VI) collagen α chain, suggesting that the cleavage product of the α3(VI) collagen does not elicit essential functions in normal growth and development. The Col6a3 mouse mutant lacking functional α3(VI) collagen chains thus serves as an animal model for COL6A3-related muscular dystrophy.


Subject(s)
Collagen Type VI/deficiency , Collagen Type VI/genetics , Muscle, Skeletal/metabolism , Tendons/metabolism , Animals , Collagen Type VI/physiology , Disease Models, Animal , Extracellular Matrix/metabolism , Genotype , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microfibrils/metabolism , Muscle, Skeletal/physiopathology , Mutation , Phenotype , Tendons/physiopathology
14.
PLoS One ; 8(2): e56716, 2013.
Article in English | MEDLINE | ID: mdl-23437220

ABSTRACT

This study identifies metabolic and protein phenotypic alterations in gastrocnemius, tibialis anterior and diaphragm muscles of Col6a1(-/-) mice, a model of human collagen VI myopathies. All three muscles of Col6a1(-/-) mice show some common changes in proteins involved in metabolism, resulting in decreased glycolysis and in changes of the TCA cycle fluxes. These changes lead to a different fate of α-ketoglutarate, with production of anabolic substrates in gastrocnemius and tibialis anterior, and with lipotoxicity in diaphragm. The metabolic changes are associated with changes of proteins involved in mechanotransduction at the myotendineous junction/costameric/sarcomeric level (TN-C, FAK, ROCK1, troponin I fast) and in energy metabolism (aldolase, enolase 3, triose phosphate isomerase, creatine kinase, adenylate kinase 1, parvalbumin, IDH1 and FASN). Together, these change may explain Ca(2+) deregulation, impaired force development, increased muscle-relaxation-time and fiber damage found in the mouse model as well as in patients. The severity of these changes differs in the three muscles (gastrocnemius

Subject(s)
Calcium/metabolism , Collagen Type VI/genetics , Muscle Cells/metabolism , Muscular Diseases/metabolism , Animals , Collagen Type VI/deficiency , Collagen Type VI/metabolism , Disease Models, Animal , Humans , Mechanotransduction, Cellular , Mice , Muscle Cells/cytology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscular Diseases/genetics , Muscular Diseases/pathology , Tibia/cytology , Tibia/metabolism
15.
J Neuropathol Exp Neurol ; 71(10): 894-906, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22975586

ABSTRACT

Ullrich congenital muscular dystrophy (UCMD) is a common form of muscular dystrophy associated with defects in collagen VI. It is characterized by loss of individual muscle fibers and muscle mass and proliferation of connective and adipose tissues. We sought to investigate the mechanisms by which collagen VI regulates muscle cell survival, size, and regeneration and, in particular, the potential role of the ubiquitin-proteasome and calpain-proteolytic systems. We studied muscle biopsies of UCMD (n = 6), other myopathy (n = 12), and control patients (n = 10) and found reduced expression of atrogin-1, MURF1, and calpain-3 mRNAs in UCMD cases. Downregulation of calpain-3 was associated with changes in the nuclear immunolocalization of nuclear factor-κB. We also observed increased expression versus controls of regeneration markers at the protein and RNA levels. Satellite cell numbers did not differ in collagen VI-deficient muscle versus normal nonregenerating muscle, indicating that collagen VI does not play a key role in the maintenance of the satellite cell pool. Our results indicate that alterations in calpain-3 and nuclear factor-κB signaling pathways may contribute to muscle mass loss in UCMD muscle, whereas atrogin-1 and MURF1 are not likely to play a major role.


Subject(s)
Calpain/physiology , Collagen Type VI/deficiency , Muscle Fibers, Skeletal/physiology , Muscle Proteins/physiology , Muscular Atrophy/metabolism , NF-kappa B/physiology , Regeneration/physiology , Signal Transduction/physiology , Child , Child, Preschool , Female , Humans , Male , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Muscular Atrophy/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Young Adult
16.
Tissue Cell ; 44(1): 1-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22071216

ABSTRACT

Bone consists of type I collagen as a major protein with minor various matrix proteins. Type VI collagen is one of bone matrix proteins but its function is not known. We therefore examined the effects of type VI collagen deficiency on bone. 3D-µCT analysis revealed that type VI collagen deficiency reduced cancellous bone mass. Cortical bone mass was not affected. Type VI collagen deficiency distorted the shape of osteoblasts both in the cancellous bone and in the cambium layer of periosteal region. Furthermore, type VI collagen deficiency disorganized collagen arrangement. These data indicate that type VI collagen contributes to maintain bone mass.


Subject(s)
Bone Diseases, Metabolic/genetics , Collagen Type VI/genetics , Osteoblasts/pathology , Animals , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Bone Remodeling/genetics , Bone and Bones/metabolism , Bone and Bones/pathology , Collagen Type VI/deficiency , Collagen Type VI/physiology , Extracellular Matrix/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Polarization , Osteoclasts/pathology , X-Ray Microtomography
17.
Neuromuscul Disord ; 22(2): 139-48, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22075033

ABSTRACT

Mutations in COL6A1, COL6A2 and COL6A3 genes result in collagen VI myopathies: Ullrich congenital muscular dystrophy (UCMD), Bethlem myopathy (BM) and intermediate phenotypes. At present, none of the existing diagnostic techniques for evaluating collagen VI expression is quantitative, and the detection of subtle changes in collagen VI expression remains challenging. We investigated flow cytometry analysis as a means of quantitatively measuring collagen VI in primary fibroblasts and compared this method with the standard method of fibroblast collagen VI immunohistochemical analysis. Eight UCMD and five BM molecularly confirmed patients were studied and compared to five controls. Flow cytometry analysis consistently detected a reduction of collagen VI of at least 60% in all UCMD cases. In BM cases the levels of collagen VI were variable but on average 20% less than controls. Flow cytometry analysis provides an alternative method for screening for collagen VI deficiency at the protein level in a quantitative, time and cost-effective manner.


Subject(s)
Collagen Type VI/deficiency , Flow Cytometry , Muscular Dystrophies/diagnosis , Adolescent , Adult , Child , Collagen Type VI/genetics , Collagen Type VI/metabolism , Fibroblasts/metabolism , Humans , Middle Aged , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Mutation/genetics , Young Adult
18.
Am J Pathol ; 180(3): 1145-1158, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22200614

ABSTRACT

To investigate the importance of the vascular basal lamina in tumor blood vessel morphogenesis and function, we compared vessel development, vessel function, and progression of B16F10 melanoma tumors in the brains of wild-type and collagen VI-null mice. In 7-day tumors in the absence of collagen VI, the width of the vascular basal lamina was reduced twofold. Although the ablation of collagen VI did not alter the abundance of blood vessels, a detailed analysis of the number of either pericytes or endothelial cells (or pericyte coverage of endothelial cells) showed that collagen VI-dependent defects during the assembly of the basal lamina have negative effects on both pericyte maturation and the sprouting and survival of endothelial cells. As a result of these deficits, vessel patency was reduced by 25%, and vessel leakiness was increased threefold, resulting in a 10-fold increase in tumor hypoxia along with a fourfold increase in hypoxia-inducible factor-1α expression. In 12-day collagen VI-null tumors, vascular endothelial growth factor expression was increased throughout the tumor stroma, in contrast to the predominantly vascular pattern of vascular endothelial growth factor expression in wild-type tumors. Vessel size was correspondingly reduced in 12-day collagen VI-null tumors. Overall, these vascular deficits produced a twofold decrease in tumor volume in collagen VI-null mice, confirming that collagen VI-dependent basal lamina assembly is a critical aspect of vessel development.


Subject(s)
Brain Neoplasms/blood supply , Collagen Type VI/physiology , Melanoma/blood supply , Animals , Apoptosis , Basement Membrane/pathology , Brain Neoplasms/pathology , Cell Hypoxia/physiology , Collagen Type VI/deficiency , Disease Progression , Endothelial Cells , Endothelium, Vascular/pathology , Hypoxia-Inducible Factor 1/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Necrosis , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Patency
19.
Semin Pediatr Neurol ; 18(4): 277-88, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22172424

ABSTRACT

Congenital muscular dystrophies (CMDs) are clinically and genetically heterogeneous neuromuscular disorders with onset at birth or in infancy in which the muscle biopsy is compatible with a dystrophic myopathy. In the past 10 years, knowledge of neuromuscular disorders has dramatically increased, particularly with the exponential boost of disclosing the genetic background of CMDs. This review will highlight the clinical description of the most important forms of CMD, paying particular attention to the main keys for diagnostic approach. The diagnosis of CMDs requires the concurrence of expertise in multiple specialties (neurology, morphology, genetics, neuroradiology) available in a few centers worldwide that have achieved sufficient experience with the different CMD subtypes. Currently, molecular diagnosis is of paramount importance not only for phenotype-genotype correlations, genetic and prenatal counseling, and prognosis and aspects of management, but also concerning the imminent availability of clinical trials and treatments.


Subject(s)
Muscular Dystrophies/congenital , Muscular Dystrophies/diagnosis , Collagen Type VI/deficiency , Genetic Association Studies , Humans , Laminin/genetics , Muscle, Skeletal/pathology , Muscular Dystrophies/genetics , Phenotype
20.
Autophagy ; 7(12): 1405-6, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22082869

ABSTRACT

Skeletal muscle fibers of collagen VI null (Col6a12/2) mice show signs of degeneration due to a block in autophagy, leading to the accumulation of damaged mitochondria and excessive apoptosis. Attempts to induce autophagic flux by subjecting these mutant mice to long-term or shorter bursts of physical activity are unsuccessful (see Grumati, et al., pp. 1415­23). In normal mice, the induction of autophagy in the skeletal muscles post-exercise is able to prevent the accumulation of damaged organelles and maintain cellular homeostasis. Thus, these studies provide an important connection between autophagy and exercise physiology.


Subject(s)
Autophagy , Collagen Type VI/deficiency , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Physical Conditioning, Animal , Animals
SELECTION OF CITATIONS
SEARCH DETAIL
...