Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Lab Invest ; 102(6): 581-588, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35145203

RESUMEN

Vertebrates exhibit patterned epidermis, exemplified by scales/interscales in mice tails and grooves/ridges on the human skin surface (microtopography). Although the role of spatiotemporal regulation of stem cells (SCs) has been implicated in this process, the mechanism underlying the development of such epidermal patterns is poorly understood. Here, we show that collagen XVII (COL17), a niche for epidermal SCs, helps stabilize epidermal patterns. Gene knockout and rescue experiments revealed that COL17 maintains the width of the murine tail scale epidermis independently of epidermal cell polarity. Skin regeneration after wounding was associated with slender scale epidermis, which was alleviated by overexpression of human COL17. COL17-negative skin in human junctional epidermolysis bullosa showed a distinct epidermal pattern from COL17-positive skin that resulted from revertant mosaicism. These results demonstrate that COL17 contributes to defining mouse tail scale shapes and human skin microtopography. Our study sheds light on the role of the SC niche in tissue pattern formation.


Asunto(s)
Autoantígenos , Epidermis , Colágenos no Fibrilares , Animales , Autoantígenos/genética , Epidermis/crecimiento & desarrollo , Ratones , Colágenos no Fibrilares/deficiencia , Colágenos no Fibrilares/genética , Piel , Colágeno Tipo XVII
2.
Sci Rep ; 9(1): 5878, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30971718

RESUMEN

Fusion of myoblasts into multinucleated myofibers is crucial for skeletal muscle development and regeneration. However, the mechanisms controlling this process remain to be determined. Here we identified the involvement of a new extracellular matrix protein in myoblast fusion. Collagen XXV is a transmembrane-type collagen highly transcribed during early myogenesis when primary myofibers form. Limb muscles of E12.5 and E14.5 Col25a1-/- embryos show a clear defect in the formation of multinucleated myofibers. In cell culture, the cleaved soluble extracellular domain of the collagen XXV is sufficient to promote the formation of highly multinucleated myofibers. Col25a1 is transiently expressed during myogenic differentiation and Col25a1 transcripts are down-regulated in multinucleated myofibers by a muscle-specific microRNA, miR-499. Altogether, these findings indicate that collagen XXV is required in vivo and in vitro for the fusion of myoblasts into myofibers and give further evidence that microRNAs participate to the regulation of this process.


Asunto(s)
Diferenciación Celular , Desarrollo de Músculos , Colágenos no Fibrilares/metabolismo , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Humanos , Ratones , Ratones Noqueados , MicroARNs/química , MicroARNs/genética , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Colágenos no Fibrilares/deficiencia , Colágenos no Fibrilares/genética , Ratas , Alineación de Secuencia
4.
Science ; 351(6273): aad4395, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26912707

RESUMEN

Hair thinning and loss are prominent aging phenotypes but have an unknown mechanism. We show that hair follicle stem cell (HFSC) aging causes the stepwise miniaturization of hair follicles and eventual hair loss in wild-type mice and in humans. In vivo fate analysis of HFSCs revealed that the DNA damage response in HFSCs causes proteolysis of type XVII collagen (COL17A1/BP180), a critical molecule for HFSC maintenance, to trigger HFSC aging, characterized by the loss of stemness signatures and by epidermal commitment. Aged HFSCs are cyclically eliminated from the skin through terminal epidermal differentiation, thereby causing hair follicle miniaturization. The aging process can be recapitulated by Col17a1 deficiency and prevented by the forced maintenance of COL17A1 in HFSCs, demonstrating that COL17A1 in HFSCs orchestrates the stem cell-centric aging program of the epithelial mini-organ.


Asunto(s)
Alopecia/metabolismo , Senescencia Celular/fisiología , Folículo Piloso/patología , Colágenos no Fibrilares/deficiencia , Proteolisis , Células Madre/patología , Anciano , Envejecimiento/metabolismo , Envejecimiento/patología , Alopecia/genética , Alopecia/patología , Animales , Autoantígenos/genética , Diferenciación Celular , Senescencia Celular/genética , Daño del ADN , Desmosomas/metabolismo , Desmosomas/patología , Femenino , Inestabilidad Genómica , Folículo Piloso/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Colágenos no Fibrilares/genética , Células Madre/metabolismo , Colágeno Tipo XVII
6.
Exp Dermatol ; 21(8): 605-11, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22775995

RESUMEN

Collagen XVII (COL17), a transmembrane protein expressed in epidermal keratinocytes (EK), is targeted by pathogenic autoantibodies in bullous pemphigoid. Treatment of EK with anti-COL17 autoantibodies triggers the production of proinflammatory cytokines. In this study, we test the hypothesis that COL17 is involved in the regulation of the EK proinflammatory response, using IL-8 expression as the primary readout. The absence of COL17 in EK derived from a junctional epidermolysis bullosa patient or shRNA-mediated knockdown of COL17 in normal EK resulted in a dysregulation of IL-8 responses under various conditions. The COL17-deficient cells showed an abnormally high IL-8 response after treatment with lipopolysaccharide (LPS), ultraviolet-B radiation or tumor necrosis factor, but exhibited a blunted IL-8 response to phorbol 12-myristate 13-acetate exposure. Induction of COL17 expression in COL17-negative EK led to a normalization of the LPS-induced proinflammatory response. Although α6ß4 integrin was found to be up-regulated in COL17-deficient EK, siRNA-mediated knockdown of the α6 and ß4 subunits revealed that COL17's effects on the LPS IL-8 response are not dependent on this integrin. In LPS-treated cells, inhibition of NF-kappa B activity in COL17-negative EK resulted in a normalization of their IL-8 response, and expression of an NF-kappa B-driven reporter was shown to be higher in COL17-deficient, compared with normal EK. These findings support the hypothesis that COL17 plays an important regulatory role in the EK proinflammatory response, acting largely via NF-kappa B. Future investigations will focus on further defining the molecular basis of this novel control network.


Asunto(s)
Autoantígenos/metabolismo , Epidermólisis Ampollosa/metabolismo , Inflamación/metabolismo , Interleucina-18/metabolismo , Queratinocitos/metabolismo , Colágenos no Fibrilares/metabolismo , Autoantígenos/genética , Línea Celular , Epidermólisis Ampollosa/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/patología , Integrina alfa6beta4/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/efectos de la radiación , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , Colágenos no Fibrilares/deficiencia , Colágenos no Fibrilares/genética , ARN Interferente Pequeño/farmacología , Acetato de Tetradecanoilforbol/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Rayos Ultravioleta , Colágeno Tipo XVII
7.
Cell Tissue Res ; 348(3): 579-88, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22457199

RESUMEN

The kidney filtration barrier consists of the capillary endothelium, the glomerular basement membrane and the slit diaphragm localized between foot processes of neighbouring podocytes. We report that collagen XVII, a transmembrane molecule known to be required for epithelial adhesion, is expressed in podocytes of normal human and mouse kidneys and in endothelial cells of the glomerular filtration barrier. Immunoelectron microscopy has revealed that collagen XVII is localized in foot processes of podocytes and in the glomerular basement membrane. Its role in kidney has been analysed in knockout mice, which survive to birth but have high neonatal mortality and skin blistering and structural abnormalities in their glomeruli. Morphometric analysis has shown increases in glomerular volume fraction and surface densities of knockout kidneys, indicating an increased glomerular amount in the cortex. Collagen XVII deficiency causes effacement of podocyte foot processes; however, major slit diaphragm disruptions have not been detected. The glomerular basement membrane is split in areas in which glomerular and endothelial basement membranes meet. Differences in the expression of collagen IV, integrins α3 or ß1, laminin α5 and nephrin have not been observed in mutant mice compared with controls. We propose that collagen XVII has a function in the attachment of podocyte foot processes to the glomerular basement membrane. It probably contributes to podocyte maturation and might have a role in glomerular filtration.


Asunto(s)
Autoantígenos/metabolismo , Membrana Basal Glomerular/metabolismo , Barrera de Filtración Glomerular/metabolismo , Colágenos no Fibrilares/metabolismo , Animales , Preescolar , Femenino , Membrana Basal Glomerular/ultraestructura , Barrera de Filtración Glomerular/ultraestructura , Humanos , Ratones , Ratones Noqueados , Colágenos no Fibrilares/deficiencia , Fenotipo , Colágeno Tipo XVII
8.
Proc Natl Acad Sci U S A ; 107(32): 14345-50, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20660747

RESUMEN

Attempts to treat congenital protein deficiencies using bone marrow-derived cells have been reported. These efforts have been based on the concepts of stem cell plasticity. However, it is considered more difficult to restore structural proteins than to restore secretory enzymes. This study aims to clarify whether bone marrow transplantation (BMT) treatment can rescue epidermolysis bullosa (EB) caused by defects in keratinocyte structural proteins. BMT treatment of adult collagen XVII (Col17) knockout mice induced donor-derived keratinocytes and Col17 expression associated with the recovery of hemidesmosomal structure and better skin manifestations, as well improving the survival rate. Both hematopoietic and mesenchymal stem cells have the potential to produce Col17 in the BMT treatment model. Furthermore, human cord blood CD34(+) cells also differentiated into keratinocytes and expressed human skin component proteins in transplanted immunocompromised (NOD/SCID/gamma(c)(null)) mice. The current conventional BMT techniques have significant potential as a systemic therapeutic approach for the treatment of human EB.


Asunto(s)
Membrana Basal/metabolismo , Trasplante de Médula Ósea/fisiología , Epidermólisis Ampollosa/terapia , Proteínas de la Membrana/biosíntesis , Animales , Autoantígenos/biosíntesis , Membrana Basal/química , Epidermis , Humanos , Queratinocitos/citología , Ratones , Ratones SCID , Colágenos no Fibrilares/biosíntesis , Colágenos no Fibrilares/deficiencia , Trasplante de Células Madre , Tasa de Supervivencia , Resultado del Tratamiento , Colágeno Tipo XVII
9.
J Immunol ; 184(4): 2166-74, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20089696

RESUMEN

Bullous pemphigoid (BP), the most common autoimmune blistering disease, is caused by autoantibodies against type XVII collagen (COL17). To establish an active stable BP animal model that demonstrates the persistent inflammatory skin lesions initiated by the anti-human COL17 Abs, we used COL17-humanized (COL17(m-/-,h+)) mice that we recently produced. First, we generated immunodeficient Rag-2(-/-)/COL17-humanized mice by crossing Rag-2(-/-) mice with COL17-humanized mice. Then, splenocytes from wild-type mice that had been immunized by grafting of human COL17-transgenic mouse skin were transferred into Rag-2(-/-)/COL17-humanized mice. The recipient mice continuously produced anti-human COL17 IgG Abs in vivo and developed blisters and erosions corresponding to clinical, histological, and immunopathological features of BP, although eosinophil infiltration, one of the characteristic histological findings observed in BP patients, was not detected in the recipients. Although the depletion of CD8(+) T cells from the immunized splenocytes was found to produce no effects in the recipients, the depletion of CD4(+) T cells as well as CD45R(+) B cells was found to inhibit the production of anti-human COL17 IgG Abs in the recipients, resulting in no apparent clinical phenotype. Furthermore, we demonstrated that cyclosporin A significantly suppressed the production of anti-human COL17 IgG Abs and prevented the development of the BP phenotype in the treated recipients. Although this model in an immunodeficient mouse does not exactly reproduce the induction mechanism of BP in human patients, this unique experimental system targeting humanized pathogenic Ag allows us to investigate ongoing autoimmune responses to human molecules in experimental animal models.


Asunto(s)
Autoantígenos/genética , Modelos Animales de Enfermedad , Colágenos no Fibrilares/genética , Penfigoide Ampolloso/genética , Penfigoide Ampolloso/inmunología , Animales , Autoanticuerpos/biosíntesis , Autoantígenos/inmunología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Femenino , Marcación de Gen/métodos , Humanos , Inmunoglobulina G/biosíntesis , Inmunofenotipificación , Antígenos Comunes de Leucocito/biosíntesis , Antígenos Comunes de Leucocito/genética , Linfopenia/genética , Linfopenia/inmunología , Linfopenia/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Colágenos no Fibrilares/deficiencia , Colágenos no Fibrilares/inmunología , Penfigoide Ampolloso/patología , Colágeno Tipo XVII
11.
Nat Med ; 13(3): 378-83, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17322897

RESUMEN

Transmissibility of characteristic lesions to experimental animals may help us understand the pathomechanism of human autoimmune disease. Here we show that human autoimmune disease can be reproduced using genetically engineered model mice. Bullous pemphigoid (BP) is the most common serious autoimmune blistering skin disease, with a considerable body of indirect evidence indicating that the underlying autoantigen is collagen XVII (COL17). Passive transfer of human BP autoantibodies into mice does not induce skin lesions, probably because of differences between humans and mice in the amino acid sequence of the COL17 pathogenic epitope. We injected human BP autoantibody into Col17-knockout mice rescued by the human ortholog. This resulted in BP-like skin lesions and a human disease phenotype. Humanization of autoantigens is a new approach to the study of human autoimmune diseases.


Asunto(s)
Autoantígenos/química , Colágenos no Fibrilares/genética , Penfigoide Ampolloso/inmunología , Animales , Autoanticuerpos/fisiología , Autoantígenos/genética , Autoantígenos/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Colágenos no Fibrilares/deficiencia , Colágeno Tipo XVII
12.
Blood ; 109(8): 3529-37, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17185466

RESUMEN

The syndrome of leukocyte adhesion deficiency (LAD) combined with a severe Glanzmann-type bleeding disorder has been recognized as a separate disease entity. The variability in clinical and cell biological terms has remained largely unclear. We present data on 9 cases from 7 unrelated families, with 3 patients being actively followed for more than 12 years. The disease entity, designated LAD-1/variant syndrome, presents early in life and consists of nonpussing infections from bacterial and fungal origin, as well as a severe bleeding tendency. This is compatible with 2 major blood cell types contributing to the clinical symptoms (ie, granulocytes and platelets). In granulocytes of the patients, we found adhesion and chemotaxis defects, as well as a defect in NADPH oxidase activity triggered by unopsonized zymosan. This last test can be used as a screening test for the syndrome. Many proteins and genes involved in adhesion and signaling, including small GTPases such as Rap1 and Rap2 as well as the major Rap activity-regulating molecules, were normally present. Moreover, Rap1 activation was intact in patients' blood cells. Defining the primary defect awaits genetic linkage analysis, which may be greatly helped by a more precise understanding and awareness of the disease combined with the early identification of affected patients.


Asunto(s)
Quimiotaxis/genética , Hemorragia/genética , Colágenos no Fibrilares/deficiencia , Transducción de Señal/genética , Autoantígenos , Infecciones Bacterianas/etiología , Infecciones Bacterianas/genética , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/patología , Plaquetas/metabolismo , Plaquetas/patología , Adhesión Celular/genética , Femenino , Estudios de Seguimiento , Granulocitos/metabolismo , Granulocitos/patología , Hemorragia/complicaciones , Hemorragia/metabolismo , Hemorragia/patología , Humanos , Masculino , Complejos Multienzimáticos/metabolismo , Micosis/etiología , Micosis/genética , Micosis/metabolismo , Micosis/patología , NADH NADPH Oxidorreductasas/metabolismo , Linaje , Síndrome , Proteínas de Unión al GTP rap/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Colágeno Tipo XVII
14.
Expert Opin Biol Ther ; 4(9): 1435-43, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15335311

RESUMEN

Easy access to the organ and identification of underlying mutations in epidermolysis bullosa (EB) facilitated the first cutaneous gene therapy experiments in vitro in the mid-1990s. The leading technology was transduction of the respective cDNA carried by a retroviral vector. Using this approach, the genotypic and phenotypic hallmark features of the recessive forms of junctional EB, which are caused by loss of function of the structural proteins laminin-5 or bullous pemphigoid antigen 2/type XVII collagen of the dermo-epidermal basement membrane zone, have been corrected in vitro and in vivo using xenograft mouse models. Recently, this approach has also been shown to be feasible for the large COL7A1 gene (mutated in dystrophic EB), applying PhiC31 integrase or lentiviral vectors. Neither of these approaches has made it into a successful Phase I study on EB patients. Therefore, alternative approaches to gene correction, including modulation of splicing, are being investigated for gene therapy in EB.


Asunto(s)
Epidermólisis Ampollosa/terapia , Terapia Genética , Animales , Autoantígenos/genética , Autoantígenos/fisiología , Proteínas Portadoras , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Proteínas del Citoesqueleto , ADN Complementario/genética , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Distonina , Epidermólisis Ampollosa/clasificación , Epidermólisis Ampollosa/genética , Heterogeneidad Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Humanos , Integrina beta4/genética , Integrina beta4/fisiología , Queratina-14 , Queratinocitos/metabolismo , Queratinocitos/trasplante , Queratinas/deficiencia , Queratinas/genética , Queratinas/fisiología , Metaloproteinasas de la Matriz/fisiología , Ratones , Ratones Desnudos , Proteínas del Tejido Nervioso , Colágenos no Fibrilares/deficiencia , Colágenos no Fibrilares/genética , Colágenos no Fibrilares/fisiología , Inhibidores de Proteasas/uso terapéutico , Empalme del ARN , ARN Catalítico/uso terapéutico , Telomerasa/genética , Telomerasa/fisiología , Kalinina , Colágeno Tipo XVII
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA