Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Circ Res ; 128(4): e46-e62, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33375813

RESUMEN

RATIONALE: Pericytes are capillary mural cells playing a role in stabilizing newly formed blood vessels during development and tissue repair. Loss of pericytes has been described in several brain disorders, and genetically induced pericyte deficiency in the brain leads to increased macromolecular leakage across the blood-brain barrier (BBB). However, the molecular details of the endothelial response to pericyte deficiency remain elusive. OBJECTIVE: To map the transcriptional changes in brain endothelial cells resulting from lack of pericyte contact at single-cell level and to correlate them with regional heterogeneities in BBB function and vascular phenotype. METHODS AND RESULTS: We reveal transcriptional, morphological, and functional consequences of pericyte absence for brain endothelial cells using a combination of methodologies, including single-cell RNA sequencing, tracer analyses, and immunofluorescent detection of protein expression in pericyte-deficient adult Pdgfbret/ret mice. We find that endothelial cells without pericyte contact retain a general BBB-specific gene expression profile, however, they acquire a venous-shifted molecular pattern and become transformed regarding the expression of numerous growth factors and regulatory proteins. Adult Pdgfbret/ret brains display ongoing angiogenic sprouting without concomitant cell proliferation providing unique insights into the endothelial tip cell transcriptome. We also reveal heterogeneous modes of pericyte-deficient BBB impairment, where hotspot leakage sites display arteriolar-shifted identity and pinpoint putative BBB regulators. By testing the causal involvement of some of these using reverse genetics, we uncover a reinforcing role for angiopoietin 2 at the BBB. CONCLUSIONS: By elucidating the complexity of endothelial response to pericyte deficiency at cellular resolution, our study provides insight into the importance of brain pericytes for endothelial arterio-venous zonation, angiogenic quiescence, and a limited set of BBB functions. The BBB-reinforcing role of ANGPT2 (angiopoietin 2) is paradoxical given its wider role as TIE2 (TEK receptor tyrosine kinase) receptor antagonist and may suggest a unique and context-dependent function of ANGPT2 in the brain.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Pericitos/citología , Animales , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/patología , Proliferación Celular , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Linfocinas/deficiencia , Linfocinas/genética , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Pericitos/metabolismo , Pericitos/patología , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Análisis de la Célula Individual , Transcriptoma
2.
Nat Med ; 24(4): 463-473, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29529015

RESUMEN

Breast tumors of the basal-like, hormone receptor-negative subtype remain an unmet clinical challenge, as there is high rate of recurrence and poor survival in patients following treatment. Coevolution of the malignant mammary epithelium and its underlying stroma instigates cancer-associated fibroblasts (CAFs) to support most, if not all, hallmarks of cancer progression. Here we delineate a previously unappreciated role for CAFs as determinants of the molecular subtype of breast cancer. We identified paracrine crosstalk between cancer cells expressing platelet-derived growth factor (PDGF)-CC and CAFs expressing the cognate receptors in human basal-like mammary carcinomas. Genetic or pharmacological intervention of PDGF-CC activity in mouse models of cancer resulted in conversion of basal-like breast cancers into a hormone receptor-positive state that enhanced sensitivity to endocrine therapy in previously resistant tumors. We conclude that specification of breast cancer to the basal-like subtype is under microenvironmental control and is therapeutically actionable.


Asunto(s)
Neoplasias de la Mama/patología , Linfocinas/metabolismo , Comunicación Paracrina , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Microambiente Tumoral , Animales , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/irrigación sanguínea , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Proliferación Celular , Células Epiteliales/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Fibrosis , Humanos , Linfocinas/deficiencia , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neovascularización Patológica/patología , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Pronóstico , Modelos de Riesgos Proporcionales , Transducción de Señal , Células del Estroma/patología , Análisis de Supervivencia , Resultado del Tratamiento
3.
J Vasc Surg ; 59(5): 1402-9.e1-4, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23856609

RESUMEN

BACKGROUND: Platelet-derived growth factor C (PDGF-C) has been reported to promote angiogenesis independently of vascular endothelial growth factor (VEGF), although its significance in postnatal angiogenesis in vivo remains poorly understood. VEGF has been employed as a major molecular tool to induce therapeutic angiogenesis. However, VEGF therapy is not very effective in models of cardiovascular diseases associated with diabetes, and the mechanisms of this phenomenon still remain to be elucidated. METHODS: We used a murine model of hind limb ischemia and of streptozotocin-induced diabetes. RESULTS: Expression of PDGF-C and its receptor PDGFR-α were markedly upregulated in ischemic limbs. Treatment with a neutralizing antibody against PDGF-C significantly impaired blood flow recovery and neovascularization after ischemia almost to the same extent as a VEGF-neutralizing antibody. Mice deficient in PDGF-C exhibited reduced blood flow recovery after ischemia compared with wild-type mice, confirming a strong proangiogenic activity of PDGF-C. Next, we injected an expression vector encoding PDGF-C into ischemic limbs. Blood flow recovery and neovascularization after ischemia were significantly improved in the groups treated with PDGF-C compared with controls. Attenuation of angiogenic responses to ischemia has been reported in patients with diabetes even after VEGF treatment, although a precise mechanism remains unknown. We hypothesized that PDGF-C might relate to the impaired angiogenesis of diabetes. We tested this hypothesis by inducing diabetes by intraperitoneal injection of streptozotocin. Expression levels of PDGF-C at baseline and after ischemia were significantly lower in limb tissues of diabetic mice than in those of control mice, whereas expression levels of other members of the PDGF family and VEGF were not changed or were even higher in diabetic mice. Introduction of VEGF complementary DNA expression plasmid vector into ischemic limbs did not improve blood flow recovery. However, these changes were effectively reversed by additional introduction of the PDGF-C complementary DNA plasmid vector. CONCLUSIONS: These results indicate that downregulation of PDGF-C expression in limb tissues of diabetic mice contributes to impaired angiogenesis and suggest that introduction of PDGF-C might be a novel strategy for therapeutic angiogenesis, especially in the diabetic state. CLINICAL RELEVANCE: Angiogenesis and arteriogenesis after ischemia are attenuated in most diabetic patients, although the precise mechanisms remain unclear. Platelet-derived growth factors (PDGFs) have a variety of functions on many cell types, and PDGF-C stimulates angiogenesis and revascularizes ischemic tissues. This study indicates the role for PDGF-C as a critical regulator of impaired angiogenesis of diabetes and suggests that PDGF-C might be a novel target for the treatment of ischemic cardiovascular diseases in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Angiopatías Diabéticas/metabolismo , Isquemia/metabolismo , Linfocinas/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Anticuerpos Neutralizantes/administración & dosificación , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/fisiopatología , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/fisiopatología , Angiopatías Diabéticas/terapia , Técnicas de Transferencia de Gen , Miembro Posterior , Isquemia/genética , Isquemia/fisiopatología , Isquemia/terapia , Linfocinas/antagonistas & inhibidores , Linfocinas/deficiencia , Linfocinas/genética , Linfocinas/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropilina-1/metabolismo , Neuropilina-2/metabolismo , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/inmunología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Recuperación de la Función , Flujo Sanguíneo Regional , Transducción de Señal , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/inmunología , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
J Cell Physiol ; 228(3): 556-562, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22806240

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the most frequently diagnosed cancers and the fourth leading cause of cancer-related death in the United States, suggesting that there is an urgent need to design novel strategies for achieving better treatment outcome of patients diagnosed with PDAC. Our previous study has shown that activation of Notch and NF-κB play a critical role in the development of PDAC in the compound K-Ras(G12D) and Ink4a/Arf deficient transgenic mice. However, the exact molecular mechanism by which mutated K-Ras and Ink4a/Arf deficiency contribute to progression of PDAC remains largely elusive. In the present study, we used multiple methods, such as real-time RT-PCR, Western blotting assay, and immunohistochemistry to gain further mechanistic insight. We found that the deletion of Ink4a/Arf in K-Ras(G12D) expressing mice led to high expression of PDGF-D signaling pathway in the tumor and tumor-derived cell line (RInk-1 cells). Furthermore, PDGF-D knock-down in RInk-1 cells resulted in the inhibition of pancreatosphere formation and down-regulation of EZH2, CD44, EpCAM, and vimentin. Moreover, we demonstrated that epithelial-mesenchymal transition (EMT) was induced in the compound mice, which is linked with aggressiveness of PDAC. In addition, we demonstrated that tumors from compound transgenic mice have higher expression of cancer stem cell (CSC) markers. These results suggest that the acquisition of EMT phenotype and induction of CSC characteristics could be linked with the aggressiveness of PDAC mediated in part through the activation of PDGF-D, signaling.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Neoplasias Pancreáticas/etiología , Proteínas Proto-Oncogénicas p21(ras)/deficiencia , Animales , Carcinoma Ductal Pancreático/etiología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Genes ras , Humanos , Linfocinas/deficiencia , Linfocinas/genética , Ratones , Ratones Transgénicos , Mutación , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fenotipo , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal
5.
Am J Pathol ; 182(1): 107-17, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23141925

RESUMEN

Platelet-derived growth factors (PDGF) are key mediators of organ fibrosis. We investigated whether PDGF-C(-/-) mice or mice treated with neutralizing PDGF-C antibodies are protected from bile duct ligation-induced liver fibrosis, and we compared the effects with those of PDGF-C deficiency or neutralization on kidney fibrosis induced by unilateral ureteral obstruction. Unexpectedly, and in contrast to kidney fibrosis, PDGF-C deficiency or antagonism did not protect from liver fibrosis or functional liver impairment. Furthermore, the hepatic infiltration of monocytes/macrophages/dendritic cells and chemokine mRNA expression (CC chemokine ligand [CCL]5, CCL2, and CC chemokine receptor 2 [CCR2]) remained unchanged. Transcript expression of PDGF ligands increased in both liver and kidney fibrosis and was not affected by neutralization of PDGF-C. In kidney fibrosis, PDGF-C deficiency or antagonism led to reduced expression and signaling of PDGF-receptor (R)-α- and PDGFR-ß-chains. In contrast, in liver fibrosis there was either no difference (PDGF-C(-/-) mice) or even an upregulation of PDGFR-ß and signaling (anti-PDGF-C group). Finally, in vitro studies in portal myofibroblasts pointed to a predominant role of PDGF-B and PDGF-D signaling in liver fibrosis. In conclusion, our study revealed significant differences between kidney and liver fibrosis in that PDGF-C mediates kidney fibrosis, whereas antagonism of PDGF-C in liver fibrosis appears to be counteracted by significant upregulation and increased PDGFR-ß signaling. PDGF-C antagonism, therefore, may not be effective to treat liver fibrosis.


Asunto(s)
Riñón/patología , Cirrosis Hepática/metabolismo , Linfocinas/fisiología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Cirrosis Hepática/etiología , Cirrosis Hepática/prevención & control , Linfocinas/antagonistas & inhibidores , Linfocinas/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/metabolismo , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología , Obstrucción Ureteral/complicaciones
6.
Am J Pathol ; 180(3): 1136-1144, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22230248

RESUMEN

Platelet-derived growth factors (PDGFs) and their tyrosine kinase receptors (PDGFRs) are known to play important roles during development of the lungs, central nervous system (CNS), and skeleton and in several diseases. PDGF-C is a ligand for the tyrosine kinase receptor PDGFRα. Mutations in the gene encoding PDGF-C have been linked to clefts of the lip and/or palate in humans, and ablation of PDGF-C in 129/Sv background mice results in death during the perinatal period. In this study, we report that ablation of PDGF-C in C57BL/6 mice results in a milder phenotype than in 129/Sv mice, and we present a phenotypic characterization of PDGF-C deficiency in the adult murine CNS. Multiple congenital defects were observed in the CNS of PDGF-C-null C57BL/6 mice, including cerebral vascular abnormalities with abnormal vascular smooth muscle cell coverage. In vivo imaging of mice deficient in PDGF-C also revealed cerebral ventricular abnormalities, such as asymmetry of the lateral ventricles and hypoplasia of the septum, reminiscent of cavum septum pellucidum in humans. We further noted that PDGF-C-deficient mice displayed a distorted ependymal lining of the lateral ventricles, and we found evidence of misplaced neurons in the ventricular lining. We conclude that PDGF-C plays a critical role in the development of normal cerebral ventricles and neuroependymal integrity as well as in normal cerebral vascularization.


Asunto(s)
Ventrículos Cerebrales/anomalías , Epéndimo/anomalías , Malformaciones Arteriovenosas Intracraneales/etiología , Linfocinas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Animales , Linfocinas/fisiología , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
7.
Development ; 135(21): 3521-30, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18832396

RESUMEN

In the early chick embryo, Pdgfa is expressed in the epiblast, outlining the migration route that mesoderm cells expressing the receptor, Pdgfralpha, follow to form somites. Both expression of a dominant-negative PDGFRalpha and depletion of endogenous PDGFRalpha ligands through injection of PDGFRalpha-Fc fragments, inhibit the migration of mesoderm cells after their ingression through the primitive streak. siRNA-mediated downregulation of Pdgfa expression in the epiblast on one side of the streak strongly blocks the migration of mesoderm cells into that side. Beads soaked in PDGFA elicit a directional attractive movement response in mesoderm cells, showing that PDGFA can provide directional information. Surprisingly, however, PDGF signalling is also required for directional movement towards other attractants, such as FGF4. PDGF signalling controls N-cadherin expression on mesoderm cells, which is required for efficient migration. PDGF signalling activates the PI3 kinase signalling pathway in vivo and activation of this pathway is required for proper N-cadherin expression.


Asunto(s)
Cadherinas/genética , Movimiento Celular , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Mesodermo/citología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Animales , Embrión de Pollo , Pollos , Regulación hacia Abajo , Activación Enzimática , Ligandos , Linfocinas/genética , Linfocinas/metabolismo , Mesodermo/metabolismo , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Línea Primitiva/citología , Línea Primitiva/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
8.
Genesis ; 45(10): 653-7, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17941048

RESUMEN

PDGF-C is a newly identified member of the platelet-derived growth factor (PDGF) family, which is involved in multiple cellular functions by signaling through PDGF receptor (PDGFR)-alphaalpha and alphabeta dimers. PDGF-C deficiency is perinatal lethal due to the formation of cleft palate. To further characterize the cellular function of PDGF-C during both embryonic and postnatal development, we have generated two conditional alleles of the Pdgf-c gene in which two loxP sites flank exon 5. Global Cre-mediated excision of the floxed exon 5 in these alleles resulted in a complete loss of PDGF-C expression and caused embryonic defects identical to those previously described for the PDGF-C null embryos. These conditional alleles will therefore be the important genetic tools for dissecting the spatial and temporal roles of PDGF-C during development and in adult tissues. Furthermore, from this work, we have also described a simple approach for creating mouse conditional alleles in an efficient manner.


Asunto(s)
Alelos , Linfocinas/genética , Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Fisura del Paladar/embriología , Cruzamientos Genéticos , Embrión de Mamíferos , Exones , Eliminación de Gen , Marcación de Gen , Ingeniería Genética , Vectores Genéticos , Heterocigoto , Integrasas , Linfocinas/deficiencia , Ratones , Ratones Noqueados , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Recombinación Genética , Células Madre/metabolismo , Transgenes
9.
Nat Genet ; 36(10): 1111-6, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15361870

RESUMEN

PDGF-C is a member of the platelet-derived growth factor (PDGF) family, which signals through PDGF receptor (PDGFR) alphaalpha and alphabeta dimers. Here we show that Pdgfc(-/-) mice die in the perinatal period owing to feeding and respiratory difficulties associated with a complete cleft of the secondary palate. This phenotype was less severe than that of Pdgfra(-/-) embryos. Pdgfc(-/-) Pdgfa(-/-) embryos developed a cleft face, subepidermal blistering, deficiency of renal cortex mesenchyme, spina bifida and skeletal and vascular defects. Complete loss of function of both ligands, therefore, phenocopied the loss of PDGFR-alpha function, suggesting that both PDGF-A and PDGF-C signal through PDGFR-alpha to regulate the development of craniofacial structures, the neural tube and mesodermal organs. Our results also show that PDGF-C signaling is a new pathway in palatogenesis, different from, and independent of, those previously implicated.


Asunto(s)
Hueso Paladar/embriología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/fisiología , Anomalías Múltiples/embriología , Anomalías Múltiples/genética , Animales , Animales Recién Nacidos , Fisura del Paladar/embriología , Fisura del Paladar/genética , Regulación del Desarrollo de la Expresión Génica , Linfocinas , Ratones , Ratones Noqueados , Fenotipo , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal , Espina Bífida Oculta/embriología , Espina Bífida Oculta/genética
10.
Am J Pathol ; 162(3): 721-9, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12598306

RESUMEN

All blood capillaries consist of endothelial tubes surrounded by mural cells referred to as pericytes. The origin, recruitment, and function of the pericytes is poorly understood, but the importance of these cells is underscored by the severe cardiovascular defects in mice genetically devoid of factors regulating pericyte recruitment to embryonic vessels, and by the association between pericyte loss and microangiopathy in diabetes mellitus. A general problem in the study of pericytes is the shortage of markers for these cells. To identify new markers for pericytes, we have taken advantage of the platelet-derived growth factor (PDGF)-B knockout mouse model, in which developing blood vessels in the central nervous system are almost completely devoid of pericytes. Using cDNA microarrays, we analyzed the gene expression in PDGF-B null embryos in comparison with corresponding wild-type embryos and searched for down-regulated genes. The most down-regulated gene present on our microarray was RGS5, a member of the RGS family of GTPase-activating proteins for G proteins. In situ hybridization identified RGS5 expression in brain pericytes, and in pericytes and vascular smooth muscle cells in certain other, but not all, locations. Absence of RGS5 expression in PDGF-B and PDGFR beta-null embryos correlated with pericyte loss in these mice. Residual RGS5 expression in rare pericytes suggested that RGS5 is a pericyte marker expressed independently of PDGF-B/R beta signaling. With RGS5 as a proof-of-principle, our data demonstrate the usefulness of microarray analysis of mouse models for abnormal pericyte development in the identification of new pericyte-specific markers.


Asunto(s)
Músculo Liso Vascular/embriología , Pericitos/citología , Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas RGS/análisis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Transcripción Genética , Animales , Becaplermina , Biomarcadores , Dermatoglifia del ADN , Embrión de Mamíferos , Femenino , Proteínas de Unión al GTP/genética , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/fisiología , Embarazo , Proteínas Proto-Oncogénicas c-sis , Proteínas RGS/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología
11.
Am J Pathol ; 161(4): 1395-407, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12368212

RESUMEN

Platelet-derived growth factor (PDGF) is a potent stimulant of smooth muscle cell migration and proliferation in culture. To test the role of PDGF in the accumulation of smooth muscle cells in vivo, we evaluated ApoE -/- mice that develop complex lesions of atherosclerosis. Fetal liver cells from PDGF-B-deficient embryos were used to replace the circulating cells of lethally irradiated ApoE -/- mice. One month after transplant, all monocytes in PDGF-B -/- chimeras are of donor origin (lack PDGF), and no PDGF-BB is detected in circulating platelets, primary sources of PDGF in lesions. Although lesion volumes are comparable in the PDGF-B +/+ and -/- chimeras at 35 weeks, lesions in PDGF-B -/- chimeras contain mostly macrophages, appear less mature, and have a reduced frequency of fibrous cap formation as compared with PDGF-B +/+ chimeras. However, after 45 weeks, smooth muscle cell accumulation in fibrous caps is indistinguishable in the two groups. Comparison of elicited peritoneal macrophages by RNase protection assay shows an altered cytokine and cytokine receptor profile in PDGF-B -/- chimeras. ApoE -/- mice were also treated for up to 50 weeks with a PDGF receptor antagonist that blocks all three PDGF receptor dimers. Blockade of the PDGF receptors similarly delays, but does not prevent, accumulation of smooth muscle and fibrous cap formation. Thus, elimination of PDGF-B from circulating cells or blockade of PDGF receptors does not appear sufficient to prevent smooth muscle accumulation in advanced lesions of atherosclerosis.


Asunto(s)
Apolipoproteínas E/genética , Plaquetas/fisiología , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/fisiología , Arteriosclerosis/genética , Arteriosclerosis/patología , Arteriosclerosis/prevención & control , Becaplermina , Plaquetas/efectos de los fármacos , Ratones , Ratones Noqueados , Piperazinas/farmacología , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/fisiología , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-sis , Quinazolinas/farmacología
12.
Exp Cell Res ; 278(1): 31-44, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12126955

RESUMEN

Mutations in the human Notch 3 gene cause the vascular stroke and dementia syndrome CADASIL (Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy) characterized by degeneration of vascular smooth muscle cells and multiple small infarcts in the white and deep gray matter of the brain. Here we have analyzed the expression pattern of the Notch 3 gene in the pre- and postnatal mouse brain. Prenatal Notch 3 expression is restricted to a scattered population of cells within the vessel wall of all major blood vessels in the developing embryo, including those that form the perineural vascular plexus. Expression in the postnatal brain is confined to a scattered cell population within the vessel wall of small to medium-sized penetrating arteries, which are the vessel type primarily affected in CADASIL patients. In contrast, no expression was observed in capillaries and veins. Notch 3 is most likely expressed in a subset of vascular smooth muscle cells, and the expression pattern of one of the Notch ligands, Serrate 1, was very similar to that observed for Notch 3. The Notch 3 expressing pattern was not significantly altered in platelet-derived growth factor B- (PDGF-B) deficient mouse embryos, demonstrating that Notch 3 expression is not under direct control of PDGF-B. These data show that Notch 3 expression is conserved between mouse and human and suggest that the mouse is a valid system for analysis of CADASIL.


Asunto(s)
Encéfalo/metabolismo , Demencia por Múltiples Infartos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Superficie Celular , Accidente Cerebrovascular/metabolismo , Animales , Animales Modificados Genéticamente , Becaplermina , Biomarcadores/análisis , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Proteínas de Unión al Calcio , Modelos Animales de Enfermedad , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Proteína Jagged-1 , Proteínas de la Membrana/genética , Ratones , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-sis , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Receptor Notch4 , Receptores Notch , Proteínas Serrate-Jagged
13.
EMBO J ; 19(9): 1998-2007, 2000 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-10790366

RESUMEN

An intracellular timer is thought to help control the timing of oligodendrocyte differentiation. We show here that the expression of the helix-loop-helix gene Id4 in oligodendrocyte precursor cells decreases in vivo and in vitro with a time course expected if Id4 is part of the timer. We also show that Id4 expression decreases prematurely when the precursor cells are induced to differentiate by mitogen withdrawal. Both Id4 mRNA and protein decrease together under all of these conditions, suggesting that the control of Id4 expression is transcriptional. Finally, we show that enforced expression of Id4 stimulates cell proliferation and blocks differentiation induced by either mitogen withdrawal or treatment with thyroid hormone. These findings suggest that a progressive fall in Id4 transcription is part of the intracellular timer that helps determine when oligodendrocyte precursor cells withdraw from the cell cycle and differentiate.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN , Secuencias Hélice-Asa-Hélice , Oligodendroglía/citología , Proteínas/química , Proteínas/metabolismo , Proteínas Represoras , Animales , Relojes Biológicos/genética , Relojes Biológicos/fisiología , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Expresión Génica , Proteína 1 Inhibidora de la Diferenciación , Proteínas Inhibidoras de la Diferenciación , Cinética , Mitógenos/deficiencia , Mitógenos/farmacología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Temperatura , Hormonas Tiroideas/farmacología , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección
14.
Development ; 124(20): 3943-53, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9374392

RESUMEN

PDGF-A(-/-) mice lack lung alveolar smooth muscle cells (SMC), exhibit reduced deposition of elastin fibres in the lung parenchyma, and develop lung emphysema due to complete failure of alveogenesis. We have mapped the expression of PDGF-A, PDGF receptor-alpha, tropoelastin, smooth muscle alpha-actin and desmin in developing lungs from wild type and PDGF-A(-/-) mice of pre- and postnatal ages in order to get insight into the mechanisms of PDGF-A-induced alveolar SMC formation and elastin deposition. PDGF-A was expressed by developing lung epithelium. Clusters of PDGF-Ralpha-positive (PDGF-Ralpha+) mesenchymal cells occurred at the distal epithelial branches until embryonic day (E) 15.5. Between E16.5 and E17.5, PDGF-Ralpha+ cells multiplied and spread to acquire positions as solitary cells in the terminal sac walls, where they remained until the onset of alveogenesis. In PDGF-A(-/-) lungs PDGF-Ralpha+ cells failed to multiply and spread and instead remained in prospective bronchiolar walls. Three phases of tropoelastin expression were seen in the developing lung, each phase characterized by a distinct pattern of expression. The third phase, tropoelastin expression by developing alveolar SMC in conjunction with alveogenesis, was specifically and completely absent in PDGF-A(-/-) lungs. We propose that lung PDGF-Ralpha+ cells are progenitors of the tropoelastin-positive alveolar SMC. We also propose that postnatal alveogenesis failure in PDGF-A(-/-) mice is due to a prenatal block in the distal spreading of PDGF-Ralpha+ cells along the tubular lung epithelium during the canalicular stage of lung development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Pulmón/embriología , Músculo Liso/citología , Músculo Liso/embriología , Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Diferenciación Celular/genética , Movimiento Celular/genética , Ratones , Ratones Noqueados , Factor de Crecimiento Derivado de Plaquetas/deficiencia
15.
Science ; 277(5323): 242-5, 1997 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-9211853

RESUMEN

Platelet-derived growth factor (PDGF)-B-deficient mouse embryos were found to lack microvascular pericytes, which normally form part of the capillary wall, and they developed numerous capillary microaneurysms that ruptured at late gestation. Endothelial cells of the sprouting capillaries in the mutant mice appeared to be unable to attract PDGF-Rbeta-positive pericyte progenitor cells. Pericytes may contribute to the mechanical stability of the capillary wall. Comparisons made between PDGF null mouse phenotypes suggest a general role for PDGFs in the development of myofibroblasts.


Asunto(s)
Aneurisma/etiología , Capilares/citología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Animales , Encéfalo/irrigación sanguínea , Capilares/embriología , Capilares/metabolismo , Movimiento Celular , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Hemorragia/etiología , Ratones , Ratones Endogámicos C57BL , Mutación , Neovascularización Fisiológica , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-sis , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas , Receptor TIE-2 , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Regulación hacia Arriba
16.
Cell ; 85(6): 863-73, 1996 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-8681381

RESUMEN

A mouse platelet-derived growth factor A chain (PDGF-A) null allele is shown to be homozygous lethal, with two distinct restriction points, one prenatally before E10 and one postnatally. Postnatally surviving PDGF-A-deficient mice develop lung emphysema secondary to the failure of alveolar septation. This is apparently caused by the loss of alveolar myofibroblasts and associated elastin fiber deposits. PDGF alpha receptor-positive cells in the lung having the location of putative alveolar myofibroblast progenitors were specifically absent in PDGF-A null mutants. We conclude that PDGF-A is crucial for alveolar myofibroblast ontogeny. We have previously shown that PDGF-B is required in the ontogeny of kidney mesangial cells. The PDGFs therefore appear to regulate the generation of specific populations of myofibroblasts during mammalian development. The two PDGF null phenotypes also reveal analogous morphogenetic functions for myofibroblast-type cells in lung and kidney organogenesis.


Asunto(s)
Factor de Crecimiento Derivado de Plaquetas/fisiología , Alveolos Pulmonares/crecimiento & desarrollo , Enfisema Pulmonar/patología , Actinas/análisis , Animales , Cardiomegalia/patología , Quimera , Cruzamientos Genéticos , Elastina/análisis , Fibroblastos/citología , Fibroblastos/patología , Marcación de Gen , Pulmón/embriología , Pulmón/ultraestructura , Ratones , Ratones Mutantes , Músculo Liso/química , Músculo Liso/citología , Fenotipo , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Alveolos Pulmonares/química , Alveolos Pulmonares/citología , Alveolos Pulmonares/patología , ARN Mensajero/análisis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Transducción de Señal/fisiología
17.
Nat Genet ; 11(1): 60-3, 1995 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7550316

RESUMEN

The aetiology of spina bifida involves genetic and environmental factors, which may be why major genes contributing to pathogenesis have not been identified. Here we report that undulated-Patch double-mutant mice have a phenotype reminiscent of an extreme form of spina bifida occulta in humans. This unexpected phenotype in double-mutant but not single-mutant mice shows that novel congenital anomalies such as spina bifida can result from interaction between products of independently segregating loci. This example of digenic inheritance may explain the often sporadic nature of spina bifida in humans.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Factor de Crecimiento Derivado de Plaquetas/fisiología , Disrafia Espinal/genética , Factores de Transcripción/fisiología , Anomalías Múltiples/embriología , Anomalías Múltiples/genética , Animales , Cruzamientos Genéticos , Quistes/genética , Proteínas de Unión al ADN/genética , Humanos , Mesodermo/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Morfogénesis/genética , Factores de Transcripción Paired Box , Trastornos de la Pigmentación/genética , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Mutación Puntual , Eliminación de Secuencia , Disrafia Espinal/embriología , Columna Vertebral/embriología , Factores de Transcripción/genética
18.
Genes Dev ; 8(16): 1875-87, 1994 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-7958863

RESUMEN

Platelet-derived growth factor (PDGF) affects the growth, migration, and function in vitro of mesenchymal cells, but little is known about its normal physiological functions in vivo. We show here that mice deficient for PDGF B die perinatally and display several anatomical and histological abnormalities. Kidney glomerular tufts do not form, apparently because of absence of mesangial cells. Instead, a single or a few distended capillary loops fill the glomerular space. The heart and some large arteries dilate in late-stage embryos. Most PDGF B mutant embryos develop fatal hemorrhages just prior to birth. Their hematological status includes erythroblastosis, macrocytic anemia, and thrombocytopenia. On the basis of these findings, we conclude that PDGF B has crucial roles in vivo in establishing certain renal and circulatory functions.


Asunto(s)
Cardiopatías Congénitas/genética , Enfermedades Hematológicas/genética , Riñón/anomalías , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Anemia/genética , Animales , Arterias/anomalías , Secuencia de Bases , Sondas de ADN/genética , Eritroblastosis Fetal/genética , Femenino , Muerte Fetal/genética , Enfermedades Hematológicas/congénito , Humanos , Recién Nacido , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Datos de Secuencia Molecular , Fenotipo , Factor de Crecimiento Derivado de Plaquetas/fisiología , Embarazo , Eliminación de Secuencia , Trombocitopenia/genética
19.
Haematologia (Budap) ; 24(4): 241-7, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1844233

RESUMEN

Platelet-Derived Growth Factor is involved in the atherogenesis and in the genesis of vascular alterations in diabetes mellitus. Its evaluation in platelet extract, by means of the mitogenic stimulation of BALB/c 3T3 fibroblasts, showed significantly reduced values in ten diabetics compared with ten normal controls. Platelet poor plasma mitogenic activity was also evaluated but showed no significant difference. The release of platelet mitogens into the circulation and a resulting acquired platelet defect are suggested. A hypothetical growth inhibitory activity can be surmised, that could both contribute to angiopathy and wound healing defect typical of diabetes mellitus.


Asunto(s)
Plaquetas/citología , Diabetes Mellitus/sangre , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Células 3T3/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Diabetes Mellitus/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Mitosis/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología
20.
Haemostasis ; 20(3): 162-8, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-2143744

RESUMEN

Mitogenic activity, measured as 3H-thymidine incorporation by NIH 3T3 cells, following stimulation with platelet-rich-plasma-derived serum (PRS), platelet-poor-plasma-derived serum and platelet extract was studied in 14 patients with myeloproliferative disorders (MPD) and 7 normal subjects. Reduced mitogenic activity was found in PRS and platelet extract of patients with MPD, as compared to controls. The average levels of platelet-derived growth factor (PDGF) equivalents were as follows: 16.3 +/- 7.2 pg/10(6) platelets in controls, 6.2 +/- 2.2 pg/10(6) (p less than 0.05) platelets in patients with polycythaemia vera, 1.8 +/- 0.4 pg/10(6) (p less than 0.01) platelets in patients with idiopathic myelofibrosis and 4.0 +/- 0.8 pg/10(6) (p less than 0.05) platelets in patients with essential thrombocythaemia (Dunnett test). A reduction of intraplatelet levels of beta-thromboglobulin, although not statistically significant, was found in the same patients. No apparent relation was found between the amount of PDGF equivalents and the degree of bone marrow fibrosis.


Asunto(s)
Trastornos Mieloproliferativos/metabolismo , Factor de Crecimiento Derivado de Plaquetas/análisis , Mielofibrosis Primaria/etiología , Adulto , Anciano , Animales , Plaquetas/análisis , Línea Celular , Replicación del ADN/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Trastornos Mieloproliferativos/complicaciones , Trastornos Mieloproliferativos/patología , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/fisiología , Mielofibrosis Primaria/patología , beta-Tromboglobulina/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...