Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 8(1): 3155, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29453333

RESUMEN

Tenomodulin (Tnmd) is a type II transmembrane glycoprotein predominantly expressed in tendons and ligaments. We found that scleraxis (Scx), a member of the Twist-family of basic helix-loop-helix transcription factors, is a transcriptional activator of Tnmd expression in tenocytes. During embryonic development, Scx expression preceded that of Tnmd. Tnmd expression was nearly absent in tendons and ligaments of Scx-deficient mice generated by transcription activator-like effector nucleases-mediated gene disruption. Tnmd mRNA levels were dramatically decreased during serial passages of rat tenocytes. Scx silencing by small interfering RNA significantly suppressed endogenous Tnmd mRNA levels in tenocytes. Mouse Tnmd contains five E-box sites in the ~1-kb 5'-flanking region. A 174-base pair genomic fragment containing a TATA box drives transcription in tenocytes. Enhancer activity was increased in the upstream region (-1030 to -295) of Tnmd in tenocytes, but not in NIH3T3 and C3H10T1/2 cells. Preferential binding of both Scx and Twist1 as a heterodimer with E12 or E47 to CAGATG or CATCTG and transactivation of the 5'-flanking region were confirmed by electrophoresis mobility shift and dual luciferase assays, respectively. Scx directly transactivates Tnmd via these E-boxes to positively regulate tenocyte differentiation and maturation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ligamentos/citología , Proteínas de la Membrana/genética , Tenocitos/metabolismo , Activación Transcripcional , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Biomarcadores/metabolismo , Ratones , Multimerización de Proteína , Estructura Cuaternaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sitio de Iniciación de la Transcripción
2.
Kidney Int ; 90(6): 1262-1273, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27591083

RESUMEN

Primary glomerulocystic kidney disease is a special form of renal cystic disorder characterized by Bowman's space dilatation in the absence of tubular cysts. ZEB2 is a SMAD-interacting transcription factor involved in Mowat-Wilson syndrome, a congenital disorder with an increased risk for kidney anomalies. Here we show that deletion of Zeb2 in mesenchyme-derived nephrons with either Pax2-cre or Six2-cre causes primary glomerulocystic kidney disease without tubular cysts in mice. Glomerulotubular junction analysis revealed many atubular glomeruli in the kidneys of Zeb2 knockout mice, which explains the presence of glomerular cysts in the absence of tubular dilatation. Gene expression analysis showed decreased expression of early proximal tubular markers in the kidneys of Zeb2 knockout mice preceding glomerular cyst formation, suggesting that defects in proximal tubule development during early nephrogenesis contribute to the formation of congenital atubular glomeruli. At the molecular level, Zeb2 deletion caused aberrant expression of Pkd1, Hnf1ß, and Glis3, three genes causing glomerular cysts. Thus, Zeb2 regulates the morphogenesis of mesenchyme-derived nephrons and is required for proximal tubule development and glomerulotubular junction formation. Our findings also suggest that ZEB2 might be a novel disease gene in patients with primary glomerular cystic disease.


Asunto(s)
Enfermedades del Sistema Nervioso Central/genética , Esmalte Dental/anomalías , Diabetes Mellitus Tipo 2/genética , Proteínas de Homeodominio/fisiología , Enfermedades Renales Quísticas/genética , Riñón/embriología , Proteínas Represoras/fisiología , Animales , Proteínas de Unión al ADN , Factor Nuclear 1-beta del Hepatocito/metabolismo , Riñón/metabolismo , Ratones Noqueados , Proteínas Represoras/metabolismo , Canales Catiónicos TRPP/metabolismo , Transactivadores/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
3.
Hum Mol Genet ; 24(22): 6390-402, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26319231

RESUMEN

Mowat-Wilson syndrome (MOWS) is caused by de novo heterozygous mutation at ZEB2 (SIP1, ZFHX1B) gene, and exhibit moderate to severe intellectual disability (ID), a characteristic facial appearance, epilepsy and other congenital anomalies. Establishing a murine MOWS model is important, not only for investigating the pathogenesis of this disease, but also for identifying compounds that may improve the symptoms. However, because the heterozygous Zeb2 knockout mouse could not be maintained as a mouse line with the inbred C57BL/6 background, it was difficult to use those mice for the study of MOWS. Here, we systematically generated de novo Zeb2 Δex7/+ mice by inducing the Zeb2 mutation in the germ cells using conditional recombination system. The de novo Zeb2 Δex7/+ mice with C57BL/6 background developed multiple defects relevant to MOWS, including craniofacial abnormalities, defective corpus callosum formation and the decreased number of parvalbumin interneurons in the cortex. In behavioral analyses, these mice showed reduced motor activity, increased anxiety and impaired sociability. Notably, during the Barnes maze test, immobile Zeb2 mutant mice were observed over repeated trials. In contrast, neither the mouse line nor the de novo Zeb2 Δex7/+ mice with the closed colony ICR background showed cranial abnormalities or reduced motor activities. These results demonstrate the advantages of using de novo Zeb2 Δex7/+ mice with the C57BL/6 background as the MOWS model. To our knowledge, this is the first time an inducible de novo mutation system has been applied to murine germline cells to produce an animal model of a human congenital disease.


Asunto(s)
Enfermedad de Hirschsprung/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Discapacidad Intelectual/genética , Microcefalia/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Síndrome de Aicardi/genética , Síndrome de Aicardi/metabolismo , Animales , Corteza Cerebral/metabolismo , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Modelos Animales de Enfermedad , Epilepsia/genética , Epilepsia/metabolismo , Facies , Femenino , Estudios de Asociación Genética , Células Germinativas , Mutación de Línea Germinal , Heterocigoto , Enfermedad de Hirschsprung/metabolismo , Humanos , Discapacidad Intelectual/metabolismo , Masculino , Trastornos Mentales/genética , Trastornos Mentales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Microcefalia/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
4.
Genesis ; 52(1): 56-67, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24243579

RESUMEN

A loss of function of SIP1 (Smad interacting protein 1) in the mouse as well as in human of Mowat-Wilson syndrome results in severe and multiple defects in neural tissue development, especially in the brain. However, no detailed expression analysis of SIP1 during brain development has been previously reported. In this study, we describe the generation of an EGFP knock-in reporter mouse for the Sip1 locus and our subsequent analysis of SIP1-EGFP fusion protein expression during brain development. SIP1-EGFP expression was observed in the pyramidal neurons of the hippocampus, the dentate gyrus, and the postmitotic neurons in the cerebral cortex. In layer 5 of the cerebral cortex, SIP1-EGFP expression was complementary to the Ctip2-expressing neurons, most of which are thought to be the cortico-spinal neurons. This suggested that SIP1-EGFP expressing cells might have the specific trajectory targets other than the spinal region. We further observed SIP1-EGFP expression in oligodendrocytes of the corpus callosum and fimbria, Bergmann glial cells of the cerebellum, the olfactory bulb, and in the serotonergic and dopaminergic neurons of the raphe nuclei in the brainstem. These findings may help to clarify the unknown roles of SIP1 in these cells and the pathoetiology of Mowat-Wilson syndrome.


Asunto(s)
Encéfalo/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Encéfalo/crecimiento & desarrollo , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Cuerpo Calloso/metabolismo , Giro Dentado/crecimiento & desarrollo , Giro Dentado/metabolismo , Facies , Técnicas de Sustitución del Gen , Genes Reporteros , Enfermedad de Hirschsprung/genética , Proteínas de Homeodominio/genética , Humanos , Discapacidad Intelectual/genética , Ratones , Ratones Endogámicos C57BL , Microcefalia/genética , Células Piramidales/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Represoras/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
5.
Dev Biol ; 332(2): 196-211, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19464280

RESUMEN

During endochondral bone formation, vascular invasion initiates the replacement of avascular cartilage by bone. We demonstrate herein that the cartilage-specific overexpression of VEGF-A(164) in mice results in the hypervascularization of soft connective tissues away from cartilage. Unexpectedly, perichondrial tissue remained avascular in addition to cartilage. Hypervascularization of tissues similarly occurred when various VEGF-A isoforms were overexpressed in the chick forelimb, but also in this case perichondrial tissue and cartilage were completely devoid of vasculature. However, following bony collar formation, anti-angiogenic properties in perichondrial tissue were lost and perichondrial angiogenesis was accelerated by VEGF-A(146), VEGF-A(166), or VEGF-A(190). Once the perichondrium was vascularized, osteoclast precursors were recruited from the circulation and the induction of MMP9 and MMP13 can be observed in parallel with the activation of TGF-beta signaling. Neither perichondrial angiogenesis nor the subsequent cartilage vascularization was found to be accelerated by the non-heparin-binding VEGF-A(122) or by the VEGF-A(166)DeltaE(162)-R(166) mutant lacking a neuropilin-binding motif. Hence, perichondrial angiogenesis is a prerequisite for subsequent cartilage vascularization and is differentially regulated by VEGF-A isoforms.


Asunto(s)
Neovascularización Fisiológica , Osteogénesis , Isoformas de Proteínas/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Secuencia de Aminoácidos , Animales , Huesos/efectos de los fármacos , Huesos/fisiología , Cartílago/efectos de los fármacos , Cartílago/fisiología , Embrión de Pollo , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/fisiología , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Embrión no Mamífero/anatomía & histología , Embrión no Mamífero/fisiología , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Proteoglicanos de Heparán Sulfato/química , Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Osteoblastos/citología , Osteoblastos/fisiología , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Péptidos/genética , Péptidos/metabolismo , Isoformas de Proteínas/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
6.
Bone ; 43(6): 1047-56, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18793763

RESUMEN

Chondromodulin-I (Chm-I) is a glycoprotein that stimulates the growth of chondrocytes and inhibits angiogenesis in vitro. Mice lacking the Chm1 gene show abnormal bone metabolism and pathological angiogenesis in cardiac valves in the mature stage although they develop normally without aberrations in endochondral bone formation during embryogenesis or in cartilage development during growth. These findings indicate that Chm-I is critical under conditions of stress such as bone repair through endochondral ossification of a fracture callus. We carried out the present study to examine the expression and role of Chm-I in bone repair using a stabilized tibial fracture model, and compared fracture healing in Chm1 knockout (Chm1(-/-)) mice with that in wild-type mice. Chm-I mRNA and protein localized in the external cartilaginous callus in the reparative phase of fracture healing. Radiological examination showed a delayed union in Chm1(-/-) mice although the fracture site was covered with both external and internal calluses. Chm1 null mutation reduced external cartilaginous callus formation as judged by marked decrease of type X collagen alpha 1 (Col10a1) expression and the total amount of cartilage matrix. Interestingly, the majority of chondrocytes in the periosteal callus failed to differentiate into mature chondrocytes in Chm1(-/-) mice, while the hypertrophic maturation of chondrocytes between the cortices was not affected. These results suggest that Chm-I is involved in hypertrophic maturation of periosteal chondrocytes. Although a direct effect of Chm-I on bones is still unclear, bony callus formation was increased while external cartilaginous callus decreased in Chm1(-/-) mice. We conclude that in the absence of Chm1, predominant primary bone healing occurs due to an indirect effect induced by reduction of cartilaginous callus rather than to a direct effect on osteogenic function, resulting in a delayed union.


Asunto(s)
Callo Óseo/fisiopatología , Curación de Fractura , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas de la Membrana/fisiología , Animales , Resorción Ósea , Inmunohistoquímica , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Cell Tissue Res ; 332(1): 111-22, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18239943

RESUMEN

Chondromodulin-I (ChM-I) and tenomodulin (TeM) are homologous angiogenesis inhibitors. We have analyzed the spatial relationships between capillary networks and the localization of these molecules during mouse and chick development. ChM-I and TeM proteins have been localized to the PECAM-1-negative avascular region: ChM-I is expressed in the avascular cartilage, whereas TeM is detectable in dense connective tissues, including tendons and ligaments. We have also examined the vasculature of chick embryos by injection with India ink and have performed in situ hybridization of the ChM-I and TeM genes. The onset of ChM-I expression is associated with chondrogenesis during mouse embryonic development. ChM-I expression is also detectable in precartilaginous or noncartilaginous avascular mesenchyme in chick embryos, including the somite, sclerotome, and heart. Hence, the expression domains of ChM-I and TeM during vertebrate development incorporate the typical avascular regions of the mesenchymal tissues.


Asunto(s)
Embrión de Mamíferos/metabolismo , Embrión no Mamífero/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Mesodermo/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cartílago/embriología , Cartílago/metabolismo , Embrión de Pollo , Colágeno Tipo II/genética , Tejido Conectivo/embriología , Tejido Conectivo/metabolismo , Expresión Génica , Corazón/embriología , Proteínas del Grupo de Alta Movilidad/genética , Ligamentos/embriología , Ligamentos/metabolismo , Proteínas de la Membrana/metabolismo , Mesodermo/embriología , Ratones , Ratones Endogámicos ICR , Sistema Musculoesquelético/embriología , Sistema Musculoesquelético/metabolismo , Miocardio/metabolismo , Notocorda/embriología , Notocorda/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Factor de Transcripción SOX9 , Columna Vertebral/embriología , Columna Vertebral/metabolismo , Tendones/embriología , Tendones/metabolismo , Factores de Transcripción/genética
8.
J Biol Chem ; 282(23): 17289-96, 2007 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-17430896

RESUMEN

The supramolecular architecture of the extracellular matrix and the disposition of its specific accessory molecules give rise to variable heterotopic signaling cues for single cells. Here we have described the successful occlusion of human fibroblast growth factor-2 (FGF-2) into the cubic inclusion bodies (FGF-2 polyhedra) of the Bombyx mori cytoplasmic polyhedrosis virus (BmCPV). The polyhedra are proteinous cubic crystals of several microns in size that are insoluble in the extracellular milieu. Purified FGF-2 polyhedra were found to stimulate proliferation and phosphorylation of p44/p42 mitogen-activated protein kinase in cultured fibroblasts. Moreover, cellular responses were blocked by a synthetic inhibitor of the FGF signaling pathway, SU5402, suggesting that FGF-2 polyhedra indeed act through FGF receptors. Furthermore, FGF-2 polyhedra retained potent growth stimulatory properties even after desiccation. We have demonstrated that BmCPV polyhedra microcrystals that occlude extracellular signaling proteins are a novel and versatile tool that can be employed to analyze cellular behavior at the single cell level.


Asunto(s)
Bombyx/virología , Factor 2 de Crecimiento de Fibroblastos/química , Nucleopoliedrovirus/química , Animales , Secuencia de Bases , Western Blotting , Línea Celular , Cristalización , Medios de Cultivo Condicionados , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Ratones , Células 3T3 NIH , Transducción de Señal , Solubilidad , Spodoptera
9.
Development ; 132(21): 4719-29, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16207754

RESUMEN

The cell population and the activity of the organizer change during the course of development. We addressed the mechanism of mouse node development via an analysis of the node/notochord enhancer (NE) of Foxa2. We first identified the core element (CE) of the enhancer, which in multimeric form drives gene expression in the node. The CE was activated in Wnt/beta-catenin-treated P19 cells with a time lag, and this activation was dependent on two separate sequence motifs within the CE. These same motifs were also required for enhancer activity in transgenic embryos. We identified the Tead family of transcription factors as binding proteins for the 3' motif. Teads and their co-factor YAP65 activated the CE in P19 cells, and binding of Tead to CE was essential for enhancer activity. Inhibition of Tead activity by repressor-modified Tead compromised NE enhancer activation and notochord development in transgenic mouse embryos. Furthermore, manipulation of Tead activity in zebrafish embryos led to altered expression of foxa2 in the embryonic shield. These results suggest that Tead activates the Foxa2 enhancer core element in the mouse node in cooperation with a second factor that binds to the 5' element, and that a similar mechanism also operates in the zebrafish shield.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Elementos de Facilitación Genéticos , Factor Nuclear 3-beta del Hepatocito/genética , Notocorda/metabolismo , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Embrión de Mamíferos , Embrión no Mamífero , Factores de Transcripción Forkhead/fisiología , Ratones , Ratones Transgénicos , Organizadores Embrionarios/crecimiento & desarrollo , Factores de Transcripción de Dominio TEA , Proteínas Wnt/fisiología , Pez Cebra
10.
Arthritis Rheum ; 50(3): 828-39, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15022325

RESUMEN

OBJECTIVE: Chondromodulin I (ChM-I), a cartilage matrix protein, promotes the growth and proteoglycan synthesis of chondrocytes. However, it also inhibits angiogenesis. Since ChM-I is expressed not only in cartilage, but also in the thymus, we investigated the modulation of T cell function by ChM-I to assess its therapeutic potential in rheumatoid arthritis (RA). METHODS: The localization of ChM-I expression in mouse thymus tissue was examined by in situ hybridization. The proliferative response of peripheral blood T cells and synovial cells obtained from patients with RA was evaluated by (3)H-thymidine incorporation assay. The effects of ChM-I were examined using recombinant human ChM-I (rHuChM-I). Modulation of the antigen-specific immune response was evaluated by the recall response of splenic T cells and the delayed-type hypersensitivity response induced in the ear of mice primed with ovalbumin (OVA). Antigen-induced arthritis (AIA) was induced in mice by injecting methylated bovine serum albumin into the ankle joints 2 weeks after the priming. RESULTS: ChM-I was expressed in the cortex of the thymus. Recombinant human ChM-I suppressed the proliferative response of mouse splenic T cells and human peripheral blood T cells stimulated with anti-CD3/CD28 antibodies, in a dose-dependent manner. Production of interleukin-2 was decreased in rHuChM-I-treated mouse CD4 T cells. Ten micrograms of rHuChM-I injected intraperitoneally into OVA-primed mice suppressed the induction of the antigen-specific immune response. Finally, rHuChM-I suppressed the development of AIA, and also suppressed the proliferation of synovial cells prepared from the joints of patients with RA. CONCLUSION: These results suggest that ChM-I suppresses T cell responses and synovial cell proliferation, implying that this cartilage matrix protein has a therapeutic potential in RA.


Asunto(s)
Inhibidores de la Angiogénesis/aislamiento & purificación , Inhibidores de la Angiogénesis/farmacología , Cartílago/química , Péptidos y Proteínas de Señalización Intercelular/aislamiento & purificación , Péptidos y Proteínas de Señalización Intercelular/farmacología , Proteínas de la Membrana/aislamiento & purificación , Proteínas de la Membrana/farmacología , Linfocitos T/citología , Adyuvantes Inmunológicos , Inhibidores de la Angiogénesis/metabolismo , Animales , Formación de Anticuerpos , Antígenos/inmunología , Artritis Experimental/inducido químicamente , Artritis Experimental/inmunología , Artritis Experimental/prevención & control , Artritis Reumatoide/tratamiento farmacológico , División Celular/efectos de los fármacos , Línea Celular , Colágeno , Epítopos , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Proteínas Recombinantes/farmacología , Albúmina Sérica Bovina , Membrana Sinovial/patología , Timo/metabolismo , Factores de Tiempo , Distribución Tisular
11.
Mol Cell Biol ; 23(2): 636-44, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12509461

RESUMEN

Chondromodulin I (ChM-I) was supposed from its limited expression in cartilage and its functions in cultured chondrocytes as a major regulator in cartilage development. Here, we generated mice deficient in ChM-I by targeted disruption of the ChM-I gene. No overt abnormality was detected in endochondral bone formation during embryogenesis and cartilage development during growth stages of ChM-I(-/-) mice. However, a significant increase in bone mineral density with lowered bone resorption with respect to formation was unexpectedly found in adult ChM-I(-/-) mice. Thus, the present study established that ChM-I is a bone remodeling factor.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas de la Membrana , Alelos , Animales , Animales Recién Nacidos , Northern Blotting , Southern Blotting , Western Blotting , Desarrollo Óseo , Células de la Médula Ósea/metabolismo , Cartílago/embriología , Células Cultivadas , Femenino , Homocigoto , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Genéticos , Osteoblastos/metabolismo , ARN Mensajero/metabolismo , Recombinación Genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Clin Calcium ; 12(3): 356-62, 2002 Mar.
Artículo en Japonés | MEDLINE | ID: mdl-15775314

RESUMEN

During endochondral bone formation, vascular invasion into the cartilaginous rudiments triggers the subsequent replacement of cartilage by bone. We have found that the cartilage-derived glycoprotein, Chondromodulin- I (ChM- I ), is involved in the anti-angiogenic property of cartilage, and that its absence creates a permissive microenvironment for vascular invasion in cartilage. Recently, we reported that a novel ChM- I related molecule, Tenomodulin (TeM), is specifically expressed in tendon, epimysium, ligaments, sclera, and cornea, all of which are hypovascular or avascular. In contrast to ChM- I that is secreted and accumulates in the extracellular matrix, TeM is expressed on the cell surface as a type II transmembrane protein. Anti-angiogenic properties of TeM and ChM- I in mesenchyme will be reviewed.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...