Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Pediatr Surg Int ; 34(2): 143-147, 2018 Feb.
Article de Anglais | MEDLINE | ID: mdl-29018955

RÉSUMÉ

BACKGROUND/AIM: Laminin-1 regulates neurite outgrowth in various neuronal cells. We have previously demonstrated that laminin-1 promotes enteric neural crest-derived cell (ENCC) migration by using Sox10-VENUS transgenic mice, in which ENCCs are labeled with a green fluorescent protein, Venus. Mice lacking the endothelin-B receptor gene, Ednrb -/- mice, are widely used as a model for Hirschsprung's disease (HD). The aim of this study was to investigate the effects of laminin-1on ENCC migration in Sox10-VENUS+/Ednrb -/- mice, a newly created HD mice model. METHODS: Fetal guts were dissected on embryonic day 12.5 (E12.5). Specimens were incubated either with, or without laminin-1 for 24 h and images were taken under a stereoscopic microscope. The length from the stomach to the wavefront of ENCC migration (L-E) and the total length of the gut (L-G) were measured. Changes in the ratio of L-E to L-G (L-E/L-G) after 24 h were calculated. RESULTS: On E12.5, the wavefront of ENCC migration in the HD gut samples was located in the midgut, whereas the wavefront of ENCC in Sox10-VENUS+/Ednrb +/+ (WT) samples had reached the hindgut. After 24 h, L-E/L-G had increased by 1.49%, from 34.97 to 36.46%, in HD gut and had increased by 1.07%, from 48.08 to 49.15%, in HD with laminin-1, suggesting there was no positive effect of laminin-1 administration on ENCC migration in HD. CONCLUSIONS: Our results suggest that laminin-1 does not have a positive effect on ENCC migration in HD mice on E12.5, in contrast to the phenomenon seen in normal mice gut specimens, where laminin-1 promotes ENCC migration during the same period. This suggests that there is an impairment in the interaction between ENCC and extracellular environmental factors, which are required for normal development of the enteric nervous system, resulting in an aganglionic colon in HD.


Sujet(s)
ADN/génétique , Système nerveux entérique/anatomopathologie , Maladie de Hirschsprung/génétique , Laminine/génétique , Crête neurale/anatomopathologie , Animaux , Différenciation cellulaire/physiologie , Mouvement cellulaire/physiologie , Cellules cultivées , Modèles animaux de maladie humaine , Système nerveux entérique/métabolisme , Régulation de l'expression des gènes , Maladie de Hirschsprung/métabolisme , Maladie de Hirschsprung/anatomopathologie , Immunohistochimie , Laminine/biosynthèse , Souris , Souris transgéniques , Crête neurale/métabolisme , Réaction de polymérisation en chaîne
2.
Cell Prolif ; 46(2): 137-45, 2013 Apr.
Article de Anglais | MEDLINE | ID: mdl-23510468

RÉSUMÉ

OBJECTIVES: Fractones are extracellular matrix structures that form a niche for neural stem cells and their immediate progeny in the subventricular zone of the lateral ventricle (SVZa), the primary neurogenic zone in the adult brain. We have previously shown that heparan sulphates (HS) associated with fractones bind fibroblast growth factor-2 (FGF-2), a powerful mitotic growth factor in the SVZa. Here, our objective was to determine whether the binding of FGF-2 to fractone-HS is implicated in the mechanism leading to cell proliferation in the SVZa. MATERIALS AND METHODS: Heparitinase-1 was intracerebroventricularly injected with FGF-2 to N-desulfate HS proteoglycans and determine whether the loss of HS and of FGF-2 binding to fractones modifies FGF-2 effect on cell proliferation. We also examined in vivo the binding of Alexa-Fluor-FGF-2 in relationship with the location of HS immunoreactivity in the SVZa. RESULTS: Heparatinase-1 drastically reduced the stimulatory effect of FGF-2 on cell proliferation in the SVZa. Alexa-Fluor-FGF-2 binding was strictly co-localized with HS immunoreactivity in fractones and adjacent vascular basement membranes in the SVZa. CONCLUSIONS: Our results demonstrate that FGF-2 requires HS to stimulate cell proliferation in the SVZa and suggest that HS associated with fractones and vascular basement membranes are responsible for activating FGF-2. Therefore, fractones and vascular basement membranes may function as a HS niche to drive cell proliferation in the adult neurogenic zone.


Sujet(s)
Prolifération cellulaire , Facteur de croissance fibroblastique de type 2/pharmacologie , Ventricules latéraux/effets des médicaments et des substances chimiques , Facteurs âges , Animaux , Membrane basale/effets des médicaments et des substances chimiques , Membrane basale/métabolisme , Numération cellulaire , Femelle , Facteur de croissance fibroblastique de type 2/administration et posologie , Colorants fluorescents/métabolisme , Protéoglycanes à sulfate d'héparane/pharmacologie , Immunohistochimie , Ventricules latéraux/cytologie , Ventricules latéraux/métabolisme , Mâle , Souris , Souris de lignée BALB C , Microscopie confocale , Cellules souches neurales/cytologie , Cellules souches neurales/effets des médicaments et des substances chimiques , Cellules souches neurales/métabolisme , Polysaccharide-lyases/administration et posologie , Polysaccharide-lyases/pharmacologie , Liaison aux protéines , Niche de cellules souches
3.
Neuroscience ; 167(2): 372-83, 2010 May 05.
Article de Anglais | MEDLINE | ID: mdl-20109533

RÉSUMÉ

Memantine is classified as an NMDA receptor antagonist. We recently reported that memantine promoted the proliferation of neural progenitor cells and the production of mature granule neurons in the adult hippocampus. However, the molecular mechanism responsible for the memantine-induced promotion of cellular proliferation remains unknown. In this study we searched for a factor that mediates memantine-induced cellular proliferation, and found that pigment epithelium-derived factor (PEDF), a broad-acting neurotrophic factor, is up-regulated in the dentate gyrus of adult mice after the injection of memantine. PEDF mRNA expression increased significantly by 3.5-fold at 1 day after the injection of memantine. In addition, the expression level of PEDF protein also increased by 1.8-fold at 2 days after the injection of memantine. Immunohistochemical study using anti-PEDF antibody showed that the majority of the PEDF-expressing cells were protoplasmic and perivascular astrocytes. Using a neurosphere assay, we confirmed that PEDF enhanced cellular proliferation under the presence of fibroblast growth factor-2 (FGF-2) and epidermal growth factor (EGF) but was not involved in the multilineage potency of hippocampal progenitor cells. Over expression of PEDF by adeno-associated virus, however, did not stimulate cellular proliferation, suggesting PEDF per se does not promote cellular proliferation in vivo. These findings suggest that the memantine induced PEDF up-regulation is involved in increased proliferation of hippocampal progenitor cells.


Sujet(s)
Protéines de l'oeil/biosynthèse , Hippocampe/effets des médicaments et des substances chimiques , Mémantine/pharmacologie , Facteurs de croissance nerveuse/biosynthèse , Récepteurs du N-méthyl-D-aspartate/antagonistes et inhibiteurs , Serpines/biosynthèse , Cellules souches/effets des médicaments et des substances chimiques , Adenoviridae/génétique , Animaux , Astrocytes/cytologie , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Facteur neurotrophique dérivé du cerveau/biosynthèse , Prolifération cellulaire , Protéines de l'oeil/génétique , Facteur de croissance fibroblastique de type 2/biosynthèse , Hippocampe/cytologie , Hippocampe/métabolisme , Mâle , Souris , Souris de lignée C57BL , Facteurs de croissance nerveuse/génétique , Serpines/génétique , Cellules souches/cytologie , Cellules souches/métabolisme , Régulation positive
6.
Nat Genet ; 27(4): 431-4, 2001 Apr.
Article de Anglais | MEDLINE | ID: mdl-11279527

RÉSUMÉ

Perlecan is a large heparan sulfate (HS) proteoglycan present in all basement membranes and in some other tissues such as cartilage, and is implicated in cell growth and differentiation. Mice lacking the perlecan gene (Hspg2) have a severe chondrodysplasia with dyssegmental ossification of the spine and show radiographic, clinical and chondro-osseous morphology similar to a lethal autosomal recessive disorder in humans termed dyssegmental dysplasia, Silverman-Handmaker type (DDSH; MIM 224410). Here we report a homozygous, 89-bp duplication in exon 34 of HSPG2 in a pair of siblings with DDSH born to consanguineous parents, and heterozygous point mutations in the 5' donor site of intron 52 and in the middle of exon 73 in a third, unrelated patient, causing skipping of the entire exons 52 and 73 of the HSPG2 transcript, respectively. These mutations are predicted to cause a frameshift, resulting in a truncated protein core. The cartilage matrix from these patients stained poorly with antibody specific for perlecan, but there was staining of intracellular inclusion bodies. Biochemically, truncated perlecan was not secreted by the patient fibroblasts, but was degraded to smaller fragments within the cells. Thus, DDSH is caused by a functional null mutation of HSPG2. Our findings demonstrate the critical role of perlecan in cartilage development.


Sujet(s)
Protéoglycanes à sulfate d'héparane/génétique , Mutation , Ostéochondrodysplasies/génétique , Animaux , Protéoglycanes à sulfate d'héparane/physiologie , Humains , Nouveau-né , Mâle , Souris , Souris knockout , RT-PCR
7.
Am J Med Genet ; 106(4): 254-7, 2001.
Article de Anglais | MEDLINE | ID: mdl-11891676

RÉSUMÉ

Dyssegmental dysplasia, Silverman-Handmaker type (DDSH), is a lethal autosomal recessive form of dwarfism with characteristic anisospondylic micromelia. The remarkable similarities in the radiographic, clinical, and chondroosseous morphology of DDSH patients to those of perlecan-null mice led to the identification of mutations in the perlecan gene (HSPG2) of DDSH. Perlecan, a large heparan sulfate proteoglycan, is expressed in various tissues and is a component of all basement membrane extracellular matrices. A chondrodysplasia phenotype caused by the loss of perlecan was unexpected, because cartilage does not have basement membranes. Insertion and splicing mutations in HSPG2 of DDSH were found that were predicted to create a premature termination codon. Immunostaining and biochemical analysis revealed that the mutant perlecan molecules were unstable and not secreted into the extracellular matrix. These results indicate that DDSH is caused by functional null mutations of HSPG2 and that perlecan is essential for cartilage development. Published 2002 Wiley-Liss, Inc.


Sujet(s)
Cartilage/croissance et développement , Nanisme/métabolisme , Protéoglycanes à sulfate d'héparane/physiologie , Cartilage/métabolisme , Mutation avec décalage du cadre de lecture , Duplication de gène , Protéoglycanes à sulfate d'héparane/déficit , Humains
8.
Matrix Biol ; 19(7): 637-47, 2000 Dec.
Article de Anglais | MEDLINE | ID: mdl-11102753

RÉSUMÉ

The purpose of this study was to determine the mRNA and protein expression of laminin alpha chains at various stages of in vitro skin morphogenesis. Fibroblasts in mono-cultures express low levels of the mRNA of laminin alpha1,alpha2, alpha3 and alpha4 chains. When co-cultured with keratinocytes for 28 days, they expressed the mRNA for all these chains. Keratinocytes in monolayer expressed the laminin alpha3 chain mRNA and very low levels of the mRNA of the alpha1 and alpha2 chains, although, when recombined with fibroblasts they also expressed laminin alpha1and alpha2 mRNA, but not the laminin alpha4 mRNA. Immunocytochemistry of cells in co-culture showed that laminin alpha1, alpha3 and alpha5 chains were expressed in the epidermis, while the laminin alpha2, beta1, and gamma1 chains were noted in the dermis and at the epidermo-dermal interface. The laminin alpha1chain was first expressed during the proliferative stage (14-21 days) and the laminin alpha2 and alpha5 chains appeared later, during the differentiation stage (28-42 days). The above results suggest that epithelial-mesenchymal interactions are involved in the expression of laminin alpha chain mRNA during in vitro skin morphogenesis. In addition, there is distinct temporal and spatial expression of these chains during proliferative and differentiation stages, possibly reflecting different functions.


Sujet(s)
Laminine/génétique , Peau/métabolisme , Différenciation cellulaire , Division cellulaire , Cellules cultivées , Techniques de coculture , Fibroblastes/cytologie , Fibroblastes/métabolisme , Expression des gènes , Humains , Kératinocytes/cytologie , Kératinocytes/métabolisme , Laminine/métabolisme , Morphogenèse , ARN messager , Peau/cytologie
9.
Nat Genet ; 23(3): 354-8, 1999 Nov.
Article de Anglais | MEDLINE | ID: mdl-10545953

RÉSUMÉ

Perlecan, a large, multi-domain, heparan sulfate proteoglycan originally identified in basement membrane, interacts with extracellular matrix proteins, growth factors and receptors, and influences cellular signalling. Perlecan is present in a variety of basement membranes and in other extracellular matrix structures. We have disrupted the gene encoding perlecan (Hspg2) in mice. Approximately 40% of Hspg2-/- mice died at embryonic day (E) 10.5 with defective cephalic development. The remaining Hspg2-/- mice died just after birth with skeletal dysplasia characterized by micromelia with broad and bowed long bones, narrow thorax and craniofacial abnormalities. Only 6% of Hspg2-/- mice developed both exencephaly and chondrodysplasia. Hspg2-/- cartilage showed severe disorganization of the columnar structures of chondrocytes and defective endochondral ossification. Hspg2-/- cartilage matrix contained reduced and disorganized collagen fibrils and glycosaminoglycans, suggesting that perlecan has an important role in matrix structure. In Hspg2-/- cartilage, proliferation of chondrocytes was reduced and the prehypertrophic zone was diminished. The abnormal phenotypes of the Hspg2-/- skeleton are similar to those of thanatophoric dysplasia (TD) type I, which is caused by activating mutations in FGFR3 (refs 7, 8, 9), and to those of Fgfr3 gain-of-function mice. Our findings suggest that these molecules affect similar signalling pathways.


Sujet(s)
Malformations multiples/génétique , Cartilage/croissance et développement , Tête/croissance et développement , Protéoglycanes à sulfate d'héparane , Héparitine sulfate/génétique , Héparitine sulfate/physiologie , Protein-tyrosine kinases , Protéoglycanes/génétique , Protéoglycanes/physiologie , Malformations multiples/embryologie , Malformations multiples/métabolisme , Animaux , Animaux nouveau-nés , Cartilage/malformations , Cartilage/embryologie , Cartilage/métabolisme , Protéine oligomérique de la matrice du cartilage , Différenciation cellulaire , Division cellulaire , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Protéines de la matrice extracellulaire/analyse , Délétion de gène , Expression des gènes , Glycoprotéines/analyse , Lame épiphysaire/malformations , Lame épiphysaire/métabolisme , Lame épiphysaire/anatomopathologie , Tête/malformations , Tête/embryologie , Héparitine sulfate/déficit , Humains , Matrilines , Souris , Souris transgéniques , Mutagenèse par insertion , Protéoglycanes/déficit , ARN messager/analyse , ARN messager/génétique , Récepteur de type 3 des facteurs de croissance fibroblastique , Récepteur facteur croissance fibroblaste/déficit , Récepteur facteur croissance fibroblaste/génétique , Récepteur facteur croissance fibroblaste/physiologie , Dysplasie thanatophore/génétique
10.
J Cell Biol ; 144(1): 161-73, 1999 Jan 11.
Article de Anglais | MEDLINE | ID: mdl-9885252

RÉSUMÉ

Cartilage provides the template for endochondral ossification and is crucial for determining the length and width of the skeleton. Transgenic mice with targeted expression of recombinant cartilage-derived morphogenetic protein-1 (CDMP-1), a member of the bone morphogenetic protein family, were created to investigate the role of CDMP-1 in skeletal formation. The mice exhibited chondrodysplasia with expanded cartilage, which consists of the enlarged hypertrophic zone and the reduced proliferating chondrocyte zone. Histologically, CDMP-1 increased the number of chondroprogenitor cells and accelerated chondrocyte differentiation to hypertrophy. Expression of CDMP-1 in the notochord inhibited vertebral body formation by blocking migration of sclerotome cells to the notochord. These results indicate that CDMP-1 antagonizes the ventralization signals from the notochord. Our study suggests a molecular mechanism by which CDMP-1 regulates the formation, growth, and differentiation of the skeletal elements.


Sujet(s)
Protéines morphogénétiques osseuses , Os et tissu osseux/embryologie , Chondrocytes/cytologie , Substances de croissance/physiologie , Mésoderme/cytologie , Animaux , Cartilage/embryologie , Cartilage/métabolisme , Différenciation cellulaire , Expression des gènes , Facteur-5 de croissance et de différenciation , Substances de croissance/génétique , Humains , Souris , Souris transgéniques , Morphogenèse , Cellules souches
11.
Neuromuscul Disord ; 5(5): 429-38, 1995 Sep.
Article de Anglais | MEDLINE | ID: mdl-7496177

RÉSUMÉ

We report a 4-yr and 5-month-old boy with severe clinical features of an early-onset Duchenne muscular dystrophy, who had a very short (110 kDa) dystrophin at the sarcolemma. The patient had a large deletion (exons 2-44) of the dystrophin gene which was predicted to cause a reading frame shift. Sequence analysis of dystrophin mRNA in muscle revealed an alternatively spliced gene product from exons 1 to 51 that caused restoration of the reading frame, in addition to an mRNA corresponding to the DNA deletion. A consistent result was obtained by immunocytochemical analysis of muscle; i.e. positive staining for dystrophin at the sarcolemma using antibodies against the C-terminus, cysteine-rich region and last three of 24 repeat units of the central rod-domain, but not for the remaining antibodies for dystrophin that recognize the N-terminal and proximal rod-domains. Immunostaining for dystrophin-associated glycoproteins (DAGs: 43 and 50 K) and merosin were preserved. Utrophin staining was positive but fainter than other DMD muscles. These results suggest that an extremely short dystrophin lacking the entire actin-binding site in the N-terminus cannot function properly even if the protein possesses the putative DAG-binding cysteine-rich and the C-terminal domains, and still has an ability to associate with sarcolemmal membrane.


Sujet(s)
Protéines du cytosquelette/métabolisme , Dystrophine/métabolisme , Glycoprotéines membranaires/métabolisme , Dystrophies musculaires/métabolisme , Séquence nucléotidique , Sites de fixation , Enfant d'âge préscolaire , Protéines du cytosquelette/génétique , ADN/analyse , Dystroglycanes , Dystrophine/génétique , Dystrophine/physiologie , Technique d'immunofluorescence directe , Humains , Immunotransfert , Mâle , Glycoprotéines membranaires/génétique , Données de séquences moléculaires , Muscles/anatomopathologie , Dystrophies musculaires/génétique , Dystrophies musculaires/anatomopathologie , Cadres ouverts de lecture , Réaction de polymérisation en chaîne , ARN messager/analyse , ARN messager/métabolisme , Sarcolemme/métabolisme
12.
J Neurol Sci ; 119(1): 53-64, 1993 Oct.
Article de Anglais | MEDLINE | ID: mdl-8246011

RÉSUMÉ

To address potential involvement of muscle basal lamina and membrane cytoskeleton proteins in the etiology of non-dystrophinopathy muscular dystrophies, we examined the immunostaining intensity and distribution of laminin subunits (A, B1, B2 and M), type IV collagen, dystrophin and spectrin in skeletal muscle biopsies from 64 myopathic patients (17 Fukuyama congenital muscular dystrophy: FCMD, 13 congenital muscular dystrophy unrelated to FCMD: other CMD, 16 Duchenne muscular dystrophy: DMD, and 18 other neuromuscular diseases. In FCMD muscle, we found a significant reduction of laminin M (merosin; a striated muscle specific basal lamina-associated protein) with approximately 26% of levels seen in controls by quantitative immunofluorescence. Other CMD and DMD muscles showed less dramatic reductions (78%, 80%, respectively). The localization of laminin M was also abnormal in FCMD muscle. Laminin B1 and B2 showed abnormalities similar to those observed with laminin M, but were less marked. Laminin A was only detected in rare regenerating fibers in control biopsies, whereas it was seen around most muscle fibers in FCMD patients, and in dystrophin deficient muscle fibers from DMD patients and its carrier. Staining intensity of type IV collagen in FCMD muscle was not significantly different from the other diseases. These findings may implicate a primary or central role for the basal lamina in FCMD muscle.


Sujet(s)
Laminine/métabolisme , Dystrophies musculaires/métabolisme , Adolescent , Adulte , Anticorps monoclonaux/analyse , Anticorps monoclonaux/immunologie , Technique de Western , Membrane cellulaire/immunologie , Membrane cellulaire/métabolisme , Enfant , Enfant d'âge préscolaire , Collagène/immunologie , Collagène/métabolisme , Densitométrie , Dystrophine/immunologie , Dystrophine/métabolisme , Femelle , Fluorescéine-5-isothiocyanate , Humains , Immunoglobuline G/immunologie , Immunohistochimie , Nourrisson , Laminine/immunologie , Mâle , Muscles/anatomopathologie , Dystrophies musculaires/anatomopathologie , Grossesse , Spectrine/immunologie , Spectrine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE