Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Vasc Res ; 58(4): 252-266, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33873190

RESUMEN

PURPOSE: The retinal vasculature is heavily invested by pericytes. Small GTPase R-Ras is highly expressed in endothelial cells and pericytes, suggesting importance of this Ras homolog for the regulation of the blood vessel wall. We investigated the specific contribution of pericyte-expressed R-Ras to the development of the retinal vasculature. METHODS: The effect of R-Ras deficiency in pericytes was analyzed in pericyte-targeted conditional Rras knockout mice at birth and during the capillary plexus formation in the neonatal retina. RESULTS: The offspring of these mice frequently exhibited unilateral microphthalmia. Analyses of the developing retinal vasculature in the eyes without microphthalmia revealed excessive endothelial cell proliferation, sprouting, and branching of the capillary plexus in these animals. These vessels were structurally defective with diminished pericyte coverage and basement membrane formation. Furthermore, these vessels showed reduced VE-cadherin staining and significantly elevated plasma leakage indicating the breakdown of the blood-retinal barrier. This defect was associated with considerable macrophage infiltration in the retina. CONCLUSIONS: The normal retinal vascular development is dependent on R-Ras expression in pericytes, and the absence of it leads to unattenuated angiogenesis and significantly weakens the blood-retinal barrier. Our findings underscore the importance of R-Ras for pericyte function during the normal eye development.


Asunto(s)
Barrera Hematorretinal/metabolismo , Microftalmía/metabolismo , Neovascularización Patológica , Pericitos/metabolismo , Vasos Retinianos/metabolismo , Proteínas ras/deficiencia , Animales , Animales Recién Nacidos , Antígenos CD/metabolismo , Barrera Hematorretinal/patología , Cadherinas/metabolismo , Movimiento Celular , Proliferación Celular , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Predisposición Genética a la Enfermedad , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microftalmía/genética , Microftalmía/patología , Pericitos/patología , Fenotipo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Vasos Retinianos/patología , Proteínas ras/genética
2.
Circulation ; 138(5): 494-508, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-29626067

RESUMEN

BACKGROUND: Peripheral vascular resistance has a major impact on arterial blood pressure levels. Endothelial C-type natriuretic peptide (CNP) participates in the local regulation of vascular tone, but the target cells remain controversial. The cGMP-producing guanylyl cyclase-B (GC-B) receptor for CNP is expressed in vascular smooth muscle cells (SMCs). However, whereas endothelial cell-specific CNP knockout mice are hypertensive, mice with deletion of GC-B in vascular SMCs have unaltered blood pressure. METHODS: We analyzed whether the vasodilating response to CNP changes along the vascular tree, ie, whether the GC-B receptor is expressed in microvascular types of cells. Mice with a floxed GC-B ( Npr2) gene were interbred with Tie2-Cre or PDGF-Rß-Cre ERT2 lines to develop mice lacking GC-B in endothelial cells or in precapillary arteriolar SMCs and capillary pericytes. Intravital microscopy, invasive and noninvasive hemodynamics, fluorescence energy transfer studies of pericyte cAMP levels in situ, and renal physiology were combined to dissect whether and how CNP/GC-B/cGMP signaling modulates microcirculatory tone and blood pressure. RESULTS: Intravital microscopy studies revealed that the vasodilatatory effect of CNP increases toward small-diameter arterioles and capillaries. CNP consistently did not prevent endothelin-1-induced acute constrictions of proximal arterioles, but fully reversed endothelin effects in precapillary arterioles and capillaries. Here, the GC-B receptor is expressed both in endothelial and mural cells, ie, in pericytes. It is notable that the vasodilatatory effects of CNP were preserved in mice with endothelial GC-B deletion, but abolished in mice lacking GC-B in microcirculatory SMCs and pericytes. CNP, via GC-B/cGMP signaling, modulates 2 signaling cascades in pericytes: it activates cGMP-dependent protein kinase I to phosphorylate downstream targets such as the cytoskeleton-associated vasodilator-activated phosphoprotein, and it inhibits phosphodiesterase 3A, thereby enhancing pericyte cAMP levels. These pathways ultimately prevent endothelin-induced increases of pericyte calcium levels and pericyte contraction. Mice with deletion of GC-B in microcirculatory SMCs and pericytes have elevated peripheral resistance and chronic arterial hypertension without a change in renal function. CONCLUSIONS: Our studies indicate that endothelial CNP regulates distal arteriolar and capillary blood flow. CNP-induced GC-B/cGMP signaling in microvascular SMCs and pericytes is essential for the maintenance of normal microvascular resistance and blood pressure.


Asunto(s)
Presión Arterial/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Hipertensión/metabolismo , Microcirculación/efectos de los fármacos , Microvasos/efectos de los fármacos , Péptido Natriurético Tipo-C/farmacología , Pericitos/metabolismo , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Técnicas Biosensibles , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , GMP Cíclico/metabolismo , Células Endoteliales/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Predisposición Genética a la Enfermedad , Hipertensión/genética , Hipertensión/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/metabolismo , Microvasos/fisiopatología , Péptido Natriurético Tipo-C/metabolismo , Comunicación Paracrina/efectos de los fármacos , Fenotipo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor Natriurético Atrial/deficiencia , Receptores del Factor Natriurético Atrial/genética
3.
Cell Rep ; 10(7): 1066-81, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25704811

RESUMEN

Strategies to target angiogenesis include inhibition of the vessel-stabilizing properties of vascular pericytes. Pericyte depletion in early-stage non-hypoxic tumors suppressed nascent angiogenesis, tumor growth, and lung metastasis. In contrast, pericyte depletion in advanced-stage hypoxic tumors with pre-established vasculature resulted in enhanced intra-tumoral hypoxia, decreased tumor growth, and increased lung metastasis. Furthermore, depletion of pericytes in post-natal retinal blood vessels resulted in abnormal and leaky vasculature. Tumor transcriptome profiling and biological validation revealed that angiopoietin signaling is a key regulatory pathway associated with pericyte targeting. Indeed, pericyte targeting in established mouse tumors increased angiopoietin-2 (ANG2/Angpt2) expression. Depletion of pericytes, coupled with targeting of ANG2 signaling, restored vascular stability in multiple model systems and decreased tumor growth and metastasis. Importantly, ANGPT2 expression correlated with poor outcome in patients with breast cancer. These results emphasize the potential utility of therapeutic regimens that target pericytes and ANG2 signaling in metastatic breast cancer.


Asunto(s)
Angiopoyetina 2/metabolismo , Neoplasias de la Mama/patología , Neoplasias Pulmonares/secundario , Angiopoyetina 2/inmunología , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Antígenos/genética , Antígenos/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal , Femenino , Mesilato de Imatinib/farmacología , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neovascularización Patológica , Pericitos/citología , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteoglicanos/deficiencia , Proteoglicanos/genética , Proteoglicanos/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Retina/fisiología , Transducción de Señal/efectos de los fármacos
4.
Exp Neurol ; 264: 127-34, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25510317

RESUMEN

Fibrosis is concomitant with repair processes following injuries in the central nervous system (CNS). Pericytes are considered as an origin of fibrosis-forming cells in the CNS. Here, we examined whether platelet-derived growth factor receptor ß (PDGFRß), a well-known indispensable molecule for migration, proliferation, and survival of pericytes, was involved in the production of extracellular matrix proteins, fibronectin and collagen type I, which is crucial for fibrosis after ischemic stroke. Immunohistochemistry demonstrated induction of PDGFRß expression in vascular cells of peri-infarct areas at 3-7days in a mouse stroke model. The PDGFRß-expressing cells extended from peri-infarct areas toward the ischemic core after day 7 while expressing fibronectin and collagen type I in the infarct areas. In contrast, desmin and α-smooth muscle actin, markers of pericytes, were only expressed in vascular cells. In PDGFRß heterozygous knockout mice, the expression of fibronectin and collagen type I was attenuated at both mRNA and protein levels with an enlargement of the infarct volume after ischemic stroke compared with that in wild-type littermates. In cultured brain pericytes, the expression of PDGF-B, PDGFRß, fibronectin, and collagen type I, but not desmin, was significantly increased by serum depletion (SD). The SD-induced upregulation of fibronectin and collagen type I was suppressed by SU11652, an inhibitor of PDGFRß, while PDGF-B further increased the SD-induced upregulation. In conclusion, the expression level of PDGFRß may be a crucial determinant of fibrosis after ischemic stroke. Moreover, PDGFRß signaling participates in the production of fibronectin and collagen type I after ischemic stroke.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/genética , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Encéfalo/citología , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Indoles/farmacología , Infarto de la Arteria Cerebral Media/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Pericitos/metabolismo , Pirroles/farmacología , ARN Mensajero/metabolismo , Ratas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Factores de Tiempo
5.
Biomed Res Int ; 2013: 848265, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24319691

RESUMEN

The superior colliculus (SC), a relay nucleus in the subcortical visual pathways, is implicated in socioemotional behaviors. Homeoprotein Otx2 and ß subunit of receptors of platelet-derived growth factor (PDGFR- ß ) have been suggested to play an important role in development of the visual system and development and maturation of GABAergic neurons. Although PDGFR- ß -knockout (KO) mice displayed socio-emotional deficits associated with parvalbumin (PV-)immunoreactive (IR) neurons, their anatomical bases in the SC were unknown. In the present study, Otx2 and PV-immunolabeling in the adult mouse SC were investigated in the PDGFR- ß KO mice. Although there were no differences in distribution patterns of Otx2 and PV-IR cells between the wild type and PDGFR- ß KO mice, the mean numbers of both of the Otx2- and PV-IR cells were significantly reduced in the PDGFR- ß KO mice. Furthermore, average diameters of Otx2- and PV-IR cells were significantly reduced in the PDGFR- ß KO mice. These findings suggest that PDGFR- ß plays a critical role in the functional development of the SC through its effects on Otx2- and PV-IR cells, provided specific roles of Otx2 protein and PV-IR cells in the development of SC neurons and visual information processing, respectively.


Asunto(s)
Factores de Transcripción Otx/metabolismo , Parvalbúminas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Colículos Superiores/metabolismo , Animales , Conducta Animal/fisiología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Conducta Social , Colículos Superiores/patología , Colículos Superiores/fisiopatología , Distribución Tisular
6.
Hippocampus ; 22(6): 1371-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21997856

RESUMEN

The physiological role of platelet-derived growth factor (PDGF) in the central nervous system (CNS) synaptic function remains uncharacterized. Here we identify physiological roles of PDGF receptor-ß (PDGFR-ß) in the CNS by conditional knockout of the gene encoding it. In the hippocampus, PDGFR-ß colocalized immunohistochemically with both presynaptic synaptophysin and postsynaptic density-95 (PSD-95). In the hippocampal CA1 region, expression levels of postsynaptic proteins, including spinophilin, drebrin, and PSD-95, were significantly decreased in PDGFR-ß knockout mice, although presynaptic synaptophysin levels remained comparable to controls. Interestingly, in hippocampal CA1 pyramidal neurons, dendritic spine density in PDGFR-ß knockout mice was significantly decreased compared with that seen in wild-type mice, although spine length and number of dendritic branches remained unchanged. Consistent with these findings, impairment in hippocampal long-term potentiation (LTP) and in hippocampus-dependent memory formation were seen in PDGFR-ß knockout mice. These results suggest PDGFR-ß plays critical roles in spine morphology and memory formation in mouse brain.


Asunto(s)
Espinas Dendríticas/metabolismo , Hipocampo/metabolismo , Trastornos de la Memoria/metabolismo , Neuronas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Animales , Espinas Dendríticas/patología , Hipocampo/patología , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Ratones , Ratones Noqueados , Neuronas/patología , Distribución Aleatoria , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética
7.
PLoS One ; 6(3): e18004, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21437241

RESUMEN

Platelet-derived growth factor (PDGF) is a potent mitogen. Extensive in vivo studies of PDGF and its receptor (PDGFR) genes have reported that PDGF plays an important role in embryogenesis and development of the central nervous system (CNS). Furthermore, PDGF and the ß subunit of the PDGF receptor (PDGFR-ß) have been reported to be associated with schizophrenia and autism. However, no study has reported on the effects of PDGF deletion on mice behavior. Here we generated novel mutant mice (PDGFR-ß KO) in which PDGFR-ß was conditionally deleted in CNS neurons using the Cre/loxP system. Mice without the Cre transgene but with floxed PDGFR-ß were used as controls. Both groups of mice reached adulthood without any apparent anatomical defects. These mice were further examined by conducting several behavioral tests for spatial memory, social interaction, conditioning, prepulse inhibition, and forced swimming. The test results indicated that the PDGFR-ß KO mice show deficits in all of these areas. Furthermore, an immunohistochemical study of the PDGFR-ß KO mice brain indicated that the number of parvalbumin (calcium-binding protein)-positive (i.e., putatively γ-aminobutyric acid-ergic) neurons was low in the amygdala, hippocampus, and medial prefrontal cortex. Neurophysiological studies indicated that sensory-evoked gamma oscillation was low in the PDGFR-ß KO mice, consistent with the observed reduction in the number of parvalbumin-positive neurons. These results suggest that PDGFR-ß plays an important role in cognitive and socioemotional functions, and that deficits in this receptor may partly underlie the cognitive and socioemotional deficits observed in schizophrenic and autistic patients.


Asunto(s)
Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Emociones/fisiología , Técnicas de Inactivación de Genes , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Conducta Social , Acústica , Animales , Conducta Animal/fisiología , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Condicionamiento Psicológico , Potenciales Evocados/fisiología , Miedo/fisiología , Congelación , Ratones , Ratones Noqueados , Actividad Motora/fisiología , Neuronas/metabolismo , Nociceptores/metabolismo , Parvalbúminas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Reflejo de Sobresalto/fisiología , Natación/fisiología , Factores de Tiempo
8.
J Clin Invest ; 120(2): 472-84, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20071776

RESUMEN

PDGFR is an important target for novel anticancer therapeutics because it is overexpressed in a wide variety of malignancies. Recently, however, several anticancer drugs that inhibit PDGFR signaling have been associated with clinical heart failure. Understanding this effect of PDGFR inhibitors has been difficult because the role of PDGFR signaling in the heart remains largely unexplored. As described herein, we have found that PDGFR-beta expression and activation increase dramatically in the hearts of mice exposed to load-induced cardiac stress. In mice in which Pdgfrb was knocked out in the heart in development or in adulthood, exposure to load-induced stress resulted in cardiac dysfunction and heart failure. Mechanistically, we showed that cardiomyocyte PDGFR-beta signaling plays a vital role in stress-induced cardiac angiogenesis. Specifically, we demonstrated that cardiomyocyte PDGFR-beta was an essential upstream regulator of the stress-induced paracrine angiogenic capacity (the angiogenic potential) of cardiomyocytes. These results demonstrate that cardiomyocyte PDGFR-beta is a regulator of the compensatory cardiac response to pressure overload-induced stress. Furthermore, our findings may provide insights into the mechanism of cardiotoxicity due to anticancer PDGFR inhibitors.


Asunto(s)
Corazón/fisiopatología , Miocitos Cardíacos/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Animales , Peso Corporal , Circulación Coronaria , Corazón/anatomía & histología , Corazón/efectos de los fármacos , Corazón/fisiología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Fosforilación , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal , Volumen Sistólico , Soporte de Peso
9.
J Neurosci Res ; 88(6): 1273-84, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19998489

RESUMEN

The neuroprotective effects of platelet-derived growth factor (PDGF) and the major signaling pathways involved in these were examined using primary cultured mouse cortical neurons subjected to H(2)O(2)-induced oxidative stress. The specific function of the PDGF beta-receptor (PDGFR-beta) was examined by the selective deletion of the corresponding gene using the Cre-loxP system in vitro. In wild-type neurons, PDGF-BB enhanced the survival of these neurons and suppressed H(2)O(2)-induced caspase-3 activation. The prosurvival effect of PDGF-AA was less than that of PDGF-BB. PDGF-BB highly activated Akt, extracellular signal-regulated kinase (ERK), c-jun amino-terminal kinase (JNK) and p38. PDGF-AA activated these molecules at lesser extent than PDGF-BB. In particular, PDGF-AA induced activation of Akt was at very low level. The neuroprotective effects of PDGF-BB were antagonized by inhibitors of phosphatidylinositol 3-kinase (PI3-K), mitogen-activated protein kinase kinase (MEK), JNK and p38. The PDGFR-beta-depleted neurons showed increased vulnerability to oxidative stress, and less responsiveness to PDGF-BB-induced cytoprotection and signal activation, in which Akt activation was most strongly suppressed. After all, these results demonstrated the neuroprotective effects of PDGF and the signaling pathways involved against oxidative stress. The effects of PDGF-BB were more potent than those of PDGF-AA. This might be due to the activation and additive effects of two PDGFRs after PDGF-BB stimulation. Furthermore, the PI3-K/Akt pathway that was deduced to be preferentially activated by PDGFR-beta may explain the potent effects of PDGF-BB.


Asunto(s)
Corteza Cerebral/fisiología , Neuronas/fisiología , Estrés Oxidativo/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Apoptosis/fisiología , Becaplermina , Caspasa 3/metabolismo , Supervivencia Celular/fisiología , Células Cultivadas , Corteza Cerebral/enzimología , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-sis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal
10.
Circ Res ; 103(12): 1393-401, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18948621

RESUMEN

The epicardium plays an essential role in coronary artery formation and myocardial development, but signals controlling the development and differentiation of this tissue are not well understood. To investigate the role of platelet-derived growth factor receptor (PDGFR)beta in development of epicardial-derived vascular smooth muscle cells (VSMCs), we examined PDGFRbeta(-/-) and PDGFRbeta epicardial mutant hearts. We found that PDGFRbeta(-/-) hearts failed to form dominant coronary vessels on the ventral heart surface, had a thinned myocardium, and completely lacked coronary VSMCs (cVSMCs). This constellation of defects was consistent with a primary defect in the epicardium. To verify that these defects were specific to epicardial derivatives, we generated mice with an epicardial deletion of PDGFRbeta that resulted in reduced cVSMCs distal to the aorta. The regional absence of cVSMCs suggested that cVSMCs could arise from 2 sources, epicardial and nonepicardial, and that both were dependent on PDGFRbeta. In the absence of PDGFRbeta signaling, epicardial cells adopted an irregular actin cytoskeleton, leading to aberrant migration of epicardial cells into the myocardium in vivo. In addition, PDGF receptor stimulation promoted epicardial cell migration, and PDGFRbeta-driven phosphoinositide 3'-kinase signaling was critical for this process. Our data demonstrate that PDGFRbeta is required for the formation of 2 distinct cVSMC populations and that loss of PDGFRbeta-PI3K signaling disrupts epicardial cell migration.


Asunto(s)
Movimiento Celular/fisiología , Vasos Coronarios/fisiología , Músculo Liso Vascular/fisiología , Pericardio/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Transducción de Señal/fisiología , Animales , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Pericardio/citología , Pericardio/metabolismo , Fosfatidilinositol 3-Quinasas/deficiencia , Fosfatidilinositol 3-Quinasas/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/genética
11.
Brain Res ; 1159: 77-85, 2007 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-17573050

RESUMEN

Although platelet-derived growth factor (PDGF)-BB activates PDGF receptor-beta (PDGFR-beta) and, in turn, inhibits the glutamate N-methyl-D-aspartate (NMDA) receptor function, whether PDGF-BB modulates the CNS function mediated by another glutamate receptors, alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptors, remains poorly understood. Here we now report the inhibitory effect of PDGF-BB on the AMPA receptor function in the nucleus tractus solitarius (NTS) by using slice patch-clamp techniques. Excitatory postsynaptic currents (EPSCs) were evoked by electrical stimulation of the tractus solitarius in mouse NTS second-order neurons. EPSCs were nearly completely eliminated by CNQX but not by MK-801, implying mediation through non-NMDA receptors. PDGF-BB significantly decreased the amplitude of EPSCs without affecting the mean decay time constant. This inhibitory effect was transient and reversible after removing PDGF-BB. Furthermore, PDGF-BB significantly reduced the amplitude of AMPA-induced currents in NTS neurons, which showed that PDGF-BB could suppress the AMPA receptor-mediated excitatory input via the postsynaptic mechanism. The inhibitory effect of PDGF-BB on EPSCs was not observed in mutant mice with conditional deletion of the PDGFR-beta gene in neurons. Together, these studies suggest that the PDGF-B/PDGFR-beta axis inhibits the AMPA receptor-mediated synaptic transmission that comprises the major part of the primary afferent to the NTS second-order neuron. The detected inhibitory action may be involved in the CNS regulation of the respiratory response.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptores AMPA/fisiología , Núcleo Solitario/citología , Transmisión Sináptica/efectos de los fármacos , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Análisis de Varianza , Animales , Becaplermina , Maleato de Dizocilpina/farmacología , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Potenciales Postsinápticos Excitadores/efectos de la radiación , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp/métodos , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Tiempo de Reacción/efectos de los fármacos , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Transmisión Sináptica/fisiología , Transmisión Sináptica/efectos de la radiación
12.
FASEB J ; 20(10): 1703-5, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16807374

RESUMEN

Normal blood microvessels are lined by pericytes, which contribute to microvessel development and stability through mechanisms that are poorly understood. Pericyte deficiency has been implicated in the pathogenesis of microvascular abnormalities associated with diabetes and tumors. However, the unambiguous identification of pericytes is still a problem because of cellular heterogeneity and few available molecular markers. Here we describe an approach to identify pericyte markers based on transcription profiling of pericyte-deficient brain microvessels isolated from platelet-derived growth factor (PDGF-B)-/- and PDGF beta receptor (PDGFRbeta)-/- mouse mutants. The approach was validated by the identification of known pericyte markers among the most down-regulated genes in PDGF-B-/- and PDGFRbeta-/- microvessels. Of candidates for novel pericyte markers, we selected ATP-sensitive potassium-channel Kir6.1 (also known as Kcnj8) and sulfonylurea receptor 2, (SUR2, also known as Abcc9), both part of the same channel complex, as well as delta homologue 1 (DLK1) for in situ hybridization, which demonstrated their specific expression in brain pericytes of mouse embryos. We also show that Kir6.1 is highly expressed in pericytes in brain but undetectable in pericytes in skin and heart. The three new brain pericyte markers are signaling molecules implicated in ion transport and intercellular signaling, potentially opening new windows on pericyte function in brain microvessels.


Asunto(s)
Encéfalo/citología , Capilares/citología , Análisis por Micromatrices/métodos , Pericitos/química , Proteínas Proto-Oncogénicas c-sis/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Transportadoras de Casetes de Unión a ATP/análisis , Animales , Biomarcadores/análisis , Encéfalo/irrigación sanguínea , Proteínas de Unión al Calcio , Embrión de Mamíferos , Perfilación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular , Canales KATP , Proteínas de la Membrana/análisis , Ratones , Ratones Mutantes , Canales de Potasio de Rectificación Interna/análisis , Receptores de Droga , Proteínas Represoras/análisis , Receptores de Sulfonilureas , Distribución Tisular
13.
J Neurochem ; 98(2): 588-600, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16805849

RESUMEN

Platelet-derived growth factors (PDGFs) and PDGF receptors (PDGFRs) are widely expressed in the mammalian CNS, though their functional significance remains unclear. The corresponding null-knockout mutations are lethal. Here, we developed novel mutant mice in which the gene encoding the beta subunit of PDGFR (PDGFR-beta) was genetically deleted in CNS neurons to elucidate the role of PDGFR-beta, particularly in the post-natal stage. Our mutant mice reached adulthood without apparent anatomical defects. In the mutant brain, immunohistochemical analyses showed that PDGFR-beta detected in neurons and in the cells in the subventricular zone of the lateral ventricle in wild-type mice was depleted, but PDGFR-beta detected in blood vessels remained unaffected. The cerebral damage after cryogenic injury was severely exacerbated in the mutants compared with controls. Furthermore, TdT-mediated dUTP-biotin nick end labeling (TUNEL)-positive neuronal cell death and lesion formation in the cerebral hemisphere were extensively exacerbated in our mutant mice after direct injection of NMDA without altered NMDA receptor expression. Our results clearly demonstrate that PDGFR-beta expressed in neurons protects them from cryogenic injury and NMDA-induced excitotoxicity.


Asunto(s)
Química Encefálica/fisiología , Encéfalo/crecimiento & desarrollo , Neuronas/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Western Blotting , Encéfalo/citología , Encéfalo/patología , Daño Encefálico Crónico/genética , Daño Encefálico Crónico/patología , Cartilla de ADN , Congelación , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Proteínas de Filamentos Intermediarios/genética , Ratones , Ratones Noqueados , Mutación , N-Metilaspartato/toxicidad , Proteínas del Tejido Nervioso/genética , Nestina , Neuronas/patología , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
J Biol Chem ; 280(10): 9375-89, 2005 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-15590688

RESUMEN

This study provides new perspectives of the unique aspects of platelet-derived growth factor beta-receptor (PDGFR-beta) signaling and biological responses through the establishment of a mutant mouse strain in which two loxP sequences were inserted into the introns of PDGFR-beta genome sequences. Isolation of skin fibroblasts from the mutant mice and Cre recombinase transfection in vitro induced PDGFR-beta gene deletion (PDGFR-betaDelta/Delta). The resultant depletion of the PDGFR-beta protein significantly attenuated platelet-derived growth factor (PDGF)-BB-induced cell migration, proliferation, and protection from H2O2-induced apoptosis of the cultured PDGFR-betaDelta/Delta dermal fibroblasts. PDGF-AA and fetal bovine serum were mitogenic and anti-apoptotic but were unable to induce the migration in PDGFR-beta Delta/Delta fibroblasts. Concerning the PDGF signaling, PDGF-BB-induced phosphorylation of Akt, ERK1/2, and JNK, but not p38, decreased in PDGFR-betaDelta/Delta fibroblasts, but PDGF-AA-induced signaling was not altered. Overexpression of the phospholipid phosphatases, SHIP2 and/or PTEN, inhibited PDGF-BB-induced phosphorylation of Akt and ERK1/2 in PDGFR-betaDelta/Delta fibroblasts but did not affect that of JNK and p38. These results indicate that disruption of distinct PDGFR-beta signaling pathways in PDGFR-betaDelta/Delta dermal fibroblasts impaired their proliferation and survival, but completely inhibits migratory response, and that PDGF-BB-induced phosphorylation of Akt and ERK1/2 possibly mediated by PDGFR-alpha is regulated, at least in part, by the lipid phosphatases SHIP2 and/or PTEN. Thus, the PDGFR-beta function on dermal fibroblasts appears to be critical in PDGF-BB action for skin wound healing and is clearly distinctive from that of PDGFR-alpha in the ligand-induced biological responses and the underlying properties of cellular signaling.


Asunto(s)
Fibroblastos/fisiología , Eliminación de Gen , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Cicatrización de Heridas/fisiología , Heridas y Lesiones/patología , Animales , Cartilla de ADN , Genotipo , Humanos , Integrasas/metabolismo , Ratones , Ratones Noqueados , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Proteínas Recombinantes/metabolismo , Mapeo Restrictivo , Piel , Transfección , Heridas y Lesiones/genética
15.
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
16.
Biochem Biophys Res Commun ; 290(4): 1220-7, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11811993

RESUMEN

Tenascin-C (TNC) knockout (TNKO) mice showed reduced proliferation of mesangial cells and abnormal restoration after habu-snake venom (HSV)-induced glomerulonephritis. In this study, we examined the relationship of TNC and platelet-derived growth factor receptor (PDGFR) in glomerular mesangial cells. TNC and PDGFR-alpha and -beta transcriptions were up-regulated in wild type (WT) mice after HSV injection, but in TNKO mice PDGFR-alpha transcription was not up-regulated. Immunohistochemistry showed that PDGFR-alpha was found in mesangial areas of colocalized alpha-smooth muscle actin, but in TNKO mice it was not detectable. In vitro studies showed that the expressions of PDGFR-alpha and -beta mRNA and protein in cultured glomerular mesangial cells (GMC) of TNKO mice were lower than those in WT GMC. These results suggest that failures of both TNC and PDGFR-alpha are a candidate for abnormal restoration of TNKO mice.


Asunto(s)
Mesangio Glomerular/metabolismo , Tenascina/deficiencia , Tenascina/genética , Animales , División Celular , Venenos de Crotálidos/toxicidad , Mesangio Glomerular/patología , Glomerulonefritis/inducido químicamente , Glomerulonefritis/genética , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tenascina/metabolismo , Transcripción Genética , Trimeresurus
17.
Blood ; 97(7): 1990-8, 2001 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11264163

RESUMEN

Platelet-derived growth factor (PDGF)-B and PDGF beta-receptor (PDGFR beta) deficiency in mice is embryonic lethal and results in cardiovascular, renal, placental, and hematologic disorders. The hematologic disorders are described, and a correlation with hepatic hypocellularity is demonstrated. To explore possible causes, the colony-forming activity of fetal liver cells in vitro was assessed, and hematopoietic chimeras were demonstrated by the transplantation of mutant fetal liver cells into lethally irradiated recipients. It was found that mutant colony formation is equivalent to that of wild-type controls. Hematopoietic chimeras reconstituted with PDGF-B(-/-), PDGFR beta(-/-), or wild-type fetal liver cells show complete engraftment (greater than 98%) with donor granulocytes, monocytes, B cells, and T cells and display none of the cardiovascular or hematologic abnormalities seen in mutants. In mouse embryos, PDGF-B is expressed by vascular endothelial cells and megakaryocytes. After birth, expression is seen in macrophages and neurons. This study demonstrates that hematopoietic PDGF-B or PDGFR beta expression is not required for hematopoiesis or integrity of the cardiovascular system. It is argued that metabolic stress arising from mutant defects in the placenta, heart, or blood vessels may lead to impaired liver growth and decreased production of blood cells. The chimera models in this study will serve as valuable tools to test the role of PDGF in inflammatory and immune responses. (Blood. 2001;97:1990-1998)


Asunto(s)
Vasos Sanguíneos/anomalías , Enfermedades Fetales/genética , Hematopoyesis/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Anemia/embriología , Anemia/genética , Anemia/metabolismo , Animales , Vasos Sanguíneos/embriología , Trasplante de Médula Ósea , Desarrollo Embrionario y Fetal/genética , Eritroblastosis Fetal/genética , Eritroblastosis Fetal/metabolismo , Femenino , Enfermedades Fetales/sangre , Enfermedades Fetales/patología , Corazón Fetal/anomalías , Trasplante de Tejido Fetal , Genes Letales , Prueba de Complementación Genética , Genotipo , Trasplante de Células Madre Hematopoyéticas , Inflamación , Riñón/anomalías , Riñón/embriología , Hígado/citología , Hígado/embriología , Masculino , Megacariocitos/citología , Ratones , Ratones Noqueados , Neovascularización Fisiológica/genética , Placenta/fisiopatología , Embarazo , Proteínas Proto-Oncogénicas c-sis/deficiencia , Proteínas Proto-Oncogénicas c-sis/genética , Quimera por Radiación , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Organismos Libres de Patógenos Específicos , Estrés Fisiológico/embriología , Estrés Fisiológico/genética , Estrés Fisiológico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...