Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 159(3): 584-96, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25417109

RESUMEN

Vascular and nervous systems, two major networks in mammalian bodies, show a high degree of anatomical parallelism and functional crosstalk. During development, neurons guide and attract blood vessels, and consequently this parallelism is established. Here, we identified a noncanonical neurovascular interaction in eye development and disease. VEGFR2, a critical endothelial receptor for VEGF, was more abundantly expressed in retinal neurons than in endothelial cells, including endothelial tip cells. Genetic deletion of VEGFR2 in neurons caused misdirected angiogenesis toward neurons, resulting in abnormally increased vascular density around neurons. Further genetic experiments revealed that this misdirected angiogenesis was attributable to an excessive amount of VEGF protein around neurons caused by insufficient engulfment of VEGF by VEGFR2-deficient neurons. Moreover, absence of neuronal VEGFR2 caused misdirected regenerative angiogenesis in ischemic retinopathy. Thus, this study revealed neurovascular crosstalk and unprecedented cellular regulation of VEGF: retinal neurons titrate VEGF to limit neuronal vascularization. PAPERFLICK:


Asunto(s)
Neovascularización Fisiológica , Neuronas/metabolismo , Retina/crecimiento & desarrollo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Endocitosis , Técnicas de Sustitución del Gen , Ratones , Ratones Noqueados , Neurogénesis , Retina/metabolismo , Retina/patología , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
2.
Dev Biol ; 477: 70-84, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34015362

RESUMEN

The C-X-C chemokine receptor CXCR4 and its ligand CXCL12 play an important role in organ-specific vascular branching morphogenesis. CXCR4 is preferentially expressed by arterial endothelial cells, and local secretion of CXCL12 determines the organotypic pattern of CXCR4+ arterial branching. Previous loss-of-function studies clearly demonstrated that CXCL12-CXCR4 signaling is necessary for proper arterial branching in the developing organs such as the skin and heart. To further understand the role of CXCL12-CXCR4 signaling in organ-specific vascular development, we generated a mouse model carrying the Cre recombinase-inducible Cxcr4 transgene. Endothelial cell-specific Cxcr4 gain-of-function embryos exhibited defective vascular remodeling and formation of a hierarchical vascular branching network in the developing skin and heart. Ectopic expression of CXCR4 in venous endothelial cells, but not in lymphatic endothelial cells, caused blood-filled, enlarged lymphatic vascular phenotypes, accompanied by edema. These data suggest that CXCR4 expression is tightly regulated in endothelial cells for appropriate vascular development in an organ-specific manner.


Asunto(s)
Vasos Sanguíneos/embriología , Células Endoteliales/fisiología , Neovascularización Fisiológica/fisiología , Receptores CXCR4/fisiología , Animales , Vasos Sanguíneos/anatomía & histología , Células Endoteliales/metabolismo , Mutación con Ganancia de Función , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Receptores CXCR4/biosíntesis , Remodelación Vascular/fisiología
3.
Hum Mol Genet ; 29(20): 3350-3360, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33030203

RESUMEN

Proteus syndrome is a progressive overgrowth disorder with vascular malformations caused by mosaic expression of the AKT1 c.49G > A, p.(E17K) activating variant which was predicted to cause lethality if expressed ubiquitously. To test that hypothesis, we used the ACTB-Cre gene to activate a conditional Akt1 p.(E17K) allele in the mouse. No offspring that was heterozygous for both Cre and the conditional allele (ßA-Akt1WT/flx) was viable. Fewer than expected numbers of ßA-Akt1WT/flx embryos were seen beginning at E11.5, but a few survived until E17.5. The phenotype ranged from mild to severe, but generally ßA-Akt1WT/flx embryos had fewer visible blood vessels and more hemorrhages than their wild-type littermates, which was suggestive of a vascular abnormality. Examination of E13.5 limb skin showed a primitive capillary network with increased branching complexity and abnormal patterning compared with wild-type skin. By E15.5, wild-type skin had undergone angiogenesis and formed a hierarchical network of remodeled vessels, whereas in ßA-Akt1WT/flx embryos, the capillary network failed to remodel. Mural cell coverage of the blood vessels was also reduced in ßA-Akt1WT/flx skin compared with that of wild type. Restricting expression of Akt1E17K to endothelial, cardiac or smooth muscle cells resulted in viable offspring and remodeled vasculature and did not recapitulate the ßA-Akt1WT/flx phenotype. We conclude that ubiquitous expression of Akt1E17K suppresses remodeling and inhibits the formation of a normal skin vasculature. We postulate that this failure prevents proper circulation necessary to support the growing embryo and that it is the result of interactions of multiple cell types with increased AKT signaling.


Asunto(s)
Pérdida del Embrión/patología , Embrión de Mamíferos/patología , Neovascularización Patológica/patología , Enfermedades Vasculares Periféricas/patología , Síndrome de Proteo/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Pérdida del Embrión/etiología , Pérdida del Embrión/metabolismo , Embrión de Mamíferos/metabolismo , Femenino , Ratones , Ratones Transgénicos , Neovascularización Patológica/etiología , Neovascularización Patológica/metabolismo , Enfermedades Vasculares Periféricas/etiología , Enfermedades Vasculares Periféricas/metabolismo , Síndrome de Proteo/etiología , Síndrome de Proteo/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal
4.
Angiogenesis ; 25(2): 205-224, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34665379

RESUMEN

In mice, embryonic dermal lymphatic development is well understood and used to study gene functions in lymphangiogenesis. Notch signaling is an evolutionarily conserved pathway that modulates cell fate decisions, which has been shown to both inhibit and promote dermal lymphangiogenesis. Here, we demonstrate distinct roles for Notch4 signaling versus canonical Notch signaling in embryonic dermal lymphangiogenesis. Actively growing embryonic dermal lymphatics expressed NOTCH1, NOTCH4, and DLL4 which correlated with Notch activity. In lymphatic endothelial cells (LECs), DLL4 activation of Notch induced a subset of Notch effectors and lymphatic genes, which were distinctly regulated by Notch1 and Notch4 activation. Treatment of LECs with VEGF-A or VEGF-C upregulated Dll4 transcripts and differentially and temporally regulated the expression of Notch1 and Hes/Hey genes. Mice nullizygous for Notch4 had an increase in the closure of the lymphangiogenic fronts which correlated with reduced vessel caliber in the maturing lymphatic plexus at E14.5 and reduced branching at E16.5. Activation of Notch4 suppressed LEC migration in a wounding assay significantly more than Notch1, suggesting a dominant role for Notch4 in regulating LEC migration. Unlike Notch4 nulls, inhibition of canonical Notch signaling by expressing a dominant negative form of MAML1 (DNMAML) in Prox1+ LECs led to increased lymphatic density consistent with an increase in LEC proliferation, described for the loss of LEC Notch1. Moreover, loss of Notch4 did not affect LEC canonical Notch signaling. Thus, we propose that Notch4 signaling and canonical Notch signaling have distinct functions in the coordination of embryonic dermal lymphangiogenesis.


Asunto(s)
Linfangiogénesis , Vasos Linfáticos , Animales , Células Endoteliales/metabolismo , Linfangiogénesis/fisiología , Sistema Linfático/metabolismo , Vasos Linfáticos/metabolismo , Ratones , Receptores Notch/metabolismo , Transducción de Señal
5.
Development ; 144(15): 2730-2736, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28694255

RESUMEN

The molecular mechanism by which NSC number is controlled in the neurogenic regions of the adult brain is not fully understood but it has been shown that vascular niche signals regulate neural stem cell (NSC) quiescence and growth. Here, we have uncovered a role for soluble amyloid precursor protein (sAPP) as a vascular niche signal in the subventricular zone (SVZ) of the lateral ventricle of the adult mouse brain. sAPP suppresses NSC growth in culture. Further in vivo studies on the role of APP in regulating NSC number in the SVZ clearly demonstrate that endothelial deletion of App causes a significant increase in the number of BrdU label-retaining NSCs in the SVZ, whereas NSC/astrocyte deletion of App has no detectable effect on the NSC number. Taken together, these results suggest that endothelial APP functions as a vascular niche signal that negatively regulates NSC growth to control the NSC number in the SVZ.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Células-Madre Neurales/citología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Citometría de Flujo , Inmunohistoquímica , Ventrículos Laterales/citología , Ventrículos Laterales/metabolismo , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nicho de Células Madre/genética , Nicho de Células Madre/fisiología
6.
J Mol Cell Cardiol ; 127: 270-276, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30639412

RESUMEN

Atherosclerosis is a chronic disorder of the vessel wall. One key regulator of disease progression is lipid handling in macrophages. However, the role of macrophage mitochondrial-dependent fatty acid ß-oxidation (FAO) in atherosclerosis is not well defined. To address this, we focused on carnitine palmitoyltransferase (CPT) 1 and 2, which play an essential role in the transport of long chain fatty acids (FAs) into the mitochondria. Using conditional alleles of these mitochondrial enzymes, we have generated myeloid-specific Cpt1a and Cpt2 knockout mutants (CPT1a M-KO and CPT2 M-KO). In culture, macrophages derived from CPT1a and CPT2 M-KO mice have impaired FAO, enhanced expression of the CD36 scavenger receptor, increased uptake of oxidized low-density lipoprotein (oxLDL), and augmented transformation into cholesterol-rich foam cells. In line with these in vitro observations, in the atherosclerosis-susceptible apolipoprotein E (ApoE) KO background, CPT2 M-KO mice demonstrated augmented atherosclerosis, accompanied by increased accumulation of aortic macrophages with elevated CD36 expression. These data suggest that macrophage FAO is athero-protective and that augmenting FAO may potentially slow atherosclerotic progression.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/patología , Progresión de la Enfermedad , Ácidos Grasos/metabolismo , Macrófagos/metabolismo , Animales , Ratones , Ratones Noqueados , Células Mieloides/metabolismo , Oxidación-Reducción
7.
Dev Dyn ; 247(5): 741-753, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29493038

RESUMEN

BACKGROUND: Traditionally, the central nervous system (CNS) has been viewed as an immune-privileged environment with no lymphatic vessels. This view was partially overturned by the discovery of lymphatic vessels in the dural membrane that surrounds the brain, in contact with the interior surface of the skull. We here examine the distribution and developmental timing of these lymphatic vessels. RESULTS: Using the Prox1-GFP BAC transgenic reporter and immunostaining with antibodies to lymphatic markers LYVE-1, Prox1, and Podoplanin, we have carried out whole-mount imaging of dural lymphatic vasculature at postnatal stages. We have found that between birth and postnatal day (P) 13, lymphatic vessels extend alongside dural blood vessels from the side of the skull toward the midline. Between P13 and P20, lymphatic vessels along the transverse sinuses reach the superior sagittal sinus (SSS) and extend along the SSS toward the olfactory bulb. CONCLUSIONS: Compared with the embryonic developmental timing of lymphatic vessels in other tissues, e.g. skin, dural lymphatic vessel development is dramatically delayed. This study provides useful anatomical data for continuing investigations of the fundamental mechanisms that underlie dural lymphatic vessel development. Developmental Dynamics 247:741-753, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Vasos Linfáticos/embriología , Animales , Encéfalo/irrigación sanguínea , Meninges/embriología , Ratones , Ratones Transgénicos , Piel/embriología
8.
Dev Biol ; 411(2): 183-194, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26872874

RESUMEN

Angiogenesis, the formation of new blood vessels by remodeling and growth of pre-existing vessels, is a highly orchestrated process that requires a tight balance between pro-angiogenic and anti-angiogenic factors and the integration of their corresponding signaling networks. The family of Rho GTPases, including RhoA, Rac1, and Cdc42, play a central role in many cell biological processes that involve cytoskeletal changes and cell movement. Specifically for Rac1, we have shown that excision of Rac1 using a Tie2-Cre animal line results in embryonic lethality in midgestation (embryonic day (E) 9.5), with multiple vascular defects. However, Tie2-Cre can be also expressed during vasculogenesis, prior to angiogenesis, and is active in some hematopoietic precursors that can affect vessel formation. To circumvent these limitations, we have now conditionally deleted Rac1 in a temporally controlled and endothelial-restricted fashion using Cdh5(PAC)-iCreERT2 transgenic mice. In this highly controlled experimental in vivo system, we now show that Rac1 is required for embryonic vascular integrity and angiogenesis, and for the formation of superficial and deep vascular networks in the post-natal developing retina, the latter involving a novel specific function for Rac1 in vertical blood vessel sprouting. Aligned with these findings, we show that RAC1 is spatially involved in endothelial cell migration, invasion, and radial sprouting activities in 3D collagen matrix in vitro models. Hence, Rac1 and its downstream molecules may represent potential anti-angiogeneic therapeutic targets for the treatment of many human diseases that involve aberrant neovascularization and blood vessel overgrowth.


Asunto(s)
Células Endoteliales/citología , Regulación del Desarrollo de la Expresión Génica , Neovascularización Fisiológica , Neuropéptidos/fisiología , Retina/embriología , Vasos Retinianos/fisiología , Proteína de Unión al GTP rac1/fisiología , Alelos , Animales , Movimiento Celular , Endotelio Vascular/metabolismo , Femenino , Genes Reporteros , Genotipo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/genética , ARN Interferente Pequeño/metabolismo , Vasos Retinianos/embriología , Proteína de Unión al GTP rac1/genética
9.
Development ; 141(23): 4489-99, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25406396

RESUMEN

Vascular development of the central nervous system and blood-brain barrier (BBB) induction are closely linked processes. The role of factors that promote endothelial sprouting and vascular leak, such as vascular endothelial growth factor A, are well described, but the factors that suppress angiogenic sprouting and their impact on the BBB are poorly understood. Here, we show that integrin αVß8 activates angiosuppressive TGFß gradients in the brain, which inhibit endothelial cell sprouting. Loss of αVß8 in the brain or downstream TGFß1-TGFBR2-ALK5-Smad3 signaling in endothelial cells increases vascular sprouting, branching and proliferation, leading to vascular dysplasia and hemorrhage. Importantly, BBB function in Itgb8 mutants is intact during early stages of vascular dysgenesis before hemorrhage. By contrast, Pdgfb(ret/ret) mice, which exhibit severe BBB disruption and vascular leak due to pericyte deficiency, have comparatively normal vascular morphogenesis and do not exhibit brain hemorrhage. Our data therefore suggest that abnormal vascular sprouting and patterning, not BBB dysfunction, underlie developmental cerebral hemorrhage.


Asunto(s)
Barrera Hematoencefálica/fisiología , Encéfalo/irrigación sanguínea , Hemorragia Cerebral/etiología , Neovascularización Patológica/complicaciones , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Encéfalo/metabolismo , Recuento de Células , Células Endoteliales/fisiología , Inmunohistoquímica , Integrinas/metabolismo , Ratones , Microscopía Confocal , Factor de Crecimiento Transformador beta/metabolismo
10.
Development ; 140(18): 3903-14, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23946447

RESUMEN

Dermal lymphatic endothelial cells (LECs) emerge from the dorsolateral region of the cardinal veins within the anterior trunk to form an intricate, branched network of lymphatic vessels during embryogenesis. Multiple growth factors and receptors are required for specification and maintenance of LECs, but the mechanisms coordinating LEC movements and morphogenesis to develop three-dimensional lymphatic network architecture are not well understood. Here, we demonstrate in mice that precise LEC sprouting is a key process leading to stereotypical lymphatic network coverage throughout the developing skin, and that transforming growth factor ß (TGFß) signaling is required for LEC sprouting and proper lymphatic network patterning in vivo. We utilized a series of conditional mutants to ablate the TGFß receptors Tgfbr1 (Alk5) and Tgfbr2 in LECs. To analyze lymphatic defects, we developed a novel, whole-mount embryonic skin imaging technique to visualize sprouting lymphangiogenesis and patterning at the lymphatic network level. Loss of TGFß signaling in LECs leads to a severe reduction in local lymphangiogenic sprouting, resulting in a significant decrease in global lymphatic network branching complexity within the skin. Our results also demonstrate that TGFß signaling negatively regulates LEC proliferation during lymphatic network formation. These data suggest a dual role for TGFß signaling during lymphatic network morphogenesis in the skin, such that it enhances LEC sprouting and branching complexity while attenuating LEC proliferation.


Asunto(s)
Linfangiogénesis , Vasos Linfáticos/embriología , Vasos Linfáticos/metabolismo , Transducción de Señal , Piel/embriología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proliferación Celular , Forma de la Célula/efectos de los fármacos , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Eliminación de Gen , Humanos , Integrasas/metabolismo , Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/efectos de los fármacos , Ratones , Modelos Biológicos , Mutación/genética , Neuropilina-2/metabolismo , Especificidad de Órganos/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/efectos de los fármacos , Piel/citología , Piel/efectos de los fármacos , Piel/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Development ; 140(7): 1475-85, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23462468

RESUMEN

Anatomical congruence of peripheral nerves and blood vessels is well recognized in a variety of tissues. Their physical proximity and similar branching patterns suggest that the development of these networks might be a coordinated process. Here we show that large diameter coronary veins serve as an intermediate template for distal sympathetic axon extension in the subepicardial layer of the dorsal ventricular wall of the developing mouse heart. Vascular smooth muscle cells (VSMCs) associate with large diameter veins during angiogenesis. In vivo and in vitro experiments demonstrate that these cells mediate extension of sympathetic axons via nerve growth factor (NGF). This association enables topological targeting of axons to final targets such as large diameter coronary arteries in the deeper myocardial layer. As axons extend along veins, arterial VSMCs begin to secrete NGF, which allows axons to reach target cells. We propose a sequential mechanism in which initial axon extension in the subepicardium is governed by transient NGF expression by VSMCs as they are recruited to coronary veins; subsequently, VSMCs in the myocardium begin to express NGF as they are recruited by remodeling arteries, attracting axons toward their final targets. The proposed mechanism underlies a distinct, stereotypical pattern of autonomic innervation that is adapted to the complex tissue structure and physiology of the heart.


Asunto(s)
Vasos Coronarios/fisiología , Corazón/embriología , Corazón/inervación , Sistema Nervioso Simpático/embriología , Animales , Axones/fisiología , Células Cultivadas , Embrión de Pollo , Vasos Coronarios/embriología , Vasos Coronarios/inervación , Técnicas de Cultivo de Embriones , Embrión de Mamíferos , Ratones , Ratones Transgénicos , Modelos Biológicos , Músculo Liso Vascular/embriología , Músculo Liso Vascular/inervación , Músculo Liso Vascular/metabolismo , Pericardio/embriología , Pericardio/inervación , Sistema Nervioso Simpático/fisiología
12.
Dev Dyn ; 244(1): 56-68, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25138596

RESUMEN

BACKGROUND: In mice, the intestinal tube develops from the splanchopleure before embryonic day 9.5. Subsequent patterning of nerves and blood vessels is critical for normal digestive function. A hierarchical branching vascular network allows for efficient nutrient absorption, while the complex enteric nervous system regulates intestinal motility as well as secretion, absorption, and blood flow. Despite the well-recognized significance of these systems, the precise mechanisms by which they develop have not been clearly established in mammals. RESULTS: Using a novel whole-mount immunohistochemical protocol, we visualize the pattern of intestinal neurovascular development in mice between embryonic day 10.5 and birth. In particular, we focus on the development and remodeling of the enteric vascular plexus, the migration and organization of enteric neural crest-derived cells, and the integration of peripheral sympathetic nerves with the enteric nervous system. These correlative data lead us to hypothesize a functional interaction between migrating neural crest-derived cells and endothelial cells of the primary capillary plexus, as well as a subsequent interaction between developing peripheral autonomic nerves and differentiated neural crest-derived cells. CONCLUSIONS: These studies provide useful anatomical data for continuing investigations on the functional mechanisms underlying intestinal organogenesis.


Asunto(s)
Intestinos , Neovascularización Fisiológica/fisiología , Cresta Neural/embriología , Nervios Periféricos/embriología , Sistema Nervioso Simpático/embriología , Animales , Intestinos/irrigación sanguínea , Intestinos/embriología , Intestinos/inervación , Ratones , Cresta Neural/citología , Nervios Periféricos/citología , Sistema Nervioso Simpático/citología
13.
J Biol Chem ; 289(43): 29801-16, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25190801

RESUMEN

Chromosome 1p36 deletion syndrome is one of the most common terminal deletions observed in humans and is related to congenital heart disease (CHD). However, the 1p36 genes that contribute to heart disease have not been clearly delineated. Human CASZ1 gene localizes to 1p36 and encodes a zinc finger transcription factor. Casz1 is required for Xenopus heart ventral midline progenitor cell differentiation. Whether Casz1 plays a role during mammalian heart development is unknown. Our aim is to determine 1p36 gene CASZ1 function at regulating heart development in mammals. We generated a Casz1 knock-out mouse using Casz1-trapped embryonic stem cells. Casz1 deletion in mice resulted in abnormal heart development including hypoplasia of myocardium, ventricular septal defect, and disorganized morphology. Hypoplasia of myocardium was caused by decreased cardiomyocyte proliferation. Comparative genome-wide RNA transcriptome analysis of Casz1 depleted embryonic hearts identifies abnormal expression of genes that are critical for muscular system development and function, such as muscle contraction genes TNNI2, TNNT1, and CKM; contractile fiber gene ACTA1; and cardiac arrhythmia associated ion channel coding genes ABCC9 and CACNA1D. The transcriptional regulation of some of these genes by Casz1 was also found in cellular models. Our results showed that loss of Casz1 during mouse development led to heart defect including cardiac noncompaction and ventricular septal defect, which phenocopies 1p36 deletion syndrome related CHD. This suggests that CASZ1 is a novel 1p36 CHD gene and that the abnormal expression of cardiac morphogenesis and contraction genes induced by loss of Casz1 contributes to the heart defect.


Asunto(s)
Desarrollo Embrionario/genética , Corazón/embriología , Morfogénesis/genética , Factores de Transcripción/metabolismo , Dedos de Zinc , Animales , Ciclo Celular/genética , Proliferación Celular , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/patología , Regulación del Desarrollo de la Expresión Génica , Humanos , Sistema Linfático/anomalías , Sistema Linfático/embriología , Sistema Linfático/patología , Ratones , Modelos Biológicos , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fenotipo , Reproducibilidad de los Resultados , Sarcómeros/metabolismo , Sarcómeros/patología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
14.
J Biol Chem ; 288(17): 12232-43, 2013 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-23467409

RESUMEN

G protein-coupled receptors (GPCRs) linked to both members of the Gα12 family of heterotrimeric G proteins α subunits, Gα12 and Gα13, regulate the activation of Rho GTPases, thereby contributing to many key biological processes. Multiple Rho GEFs have been proposed to link Gα12/13 GPCRs to Rho activation, including PDZ-RhoGEF (PRG), leukemia-associated Rho GEF (LARG), p115-RhoGEF (p115), lymphoid blast crisis (Lbc), and Dbl. PRG, LARG, and p115 share the presence of a regulator of G protein signaling homology (RGS) domain. There is limited information on the biological roles of this RGS-containing family of RhoGEFs in vivo. p115-deficient mice are viable with some defects in the immune system and gastrointestinal motor dysfunctions, whereas in an initial study we showed that mice deficient for Larg are viable and resistant to salt-induced hypertension. Here, we generated knock-out mice for Prg and observed that these mice do not display any overt phenotype. However, deficiency in Prg and Larg leads to complex developmental defects and early embryonic lethality. Signaling from Gα11/q-linked GPCRs to Rho was not impaired in mouse embryonic fibroblasts defective in all three RGS-containing RhoGEFs. However, a combined lack of Prg, Larg, and p115 expression abolished signaling through Gα12/13 to Rho and thrombin-induced cell proliferation, directional migration, and nuclear signaling through JNK and p38. These findings provide evidence of an essential role for the RGS-containing RhoGEF family in signaling to Rho by Gα12/13-coupled GPCRs, which may likely play a critical role during embryonic development.


Asunto(s)
Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Receptores de Trombina/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Línea Celular , Movimiento Celular/fisiología , Proliferación Celular , Fibroblastos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Ratones , Ratones Noqueados , Receptores del Ácido Lisofosfatídico/genética , Receptores de Trombina/genética , Factores de Intercambio de Guanina Nucleótido Rho , Transducción de Señal/fisiología , Proteínas de Unión al GTP rho/genética
15.
Development ; 138(22): 4875-86, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22007135

RESUMEN

Here, we show that a novel Rspo1-Wnt-Vegfc-Vegfr3 signaling pathway plays an essential role in developmental angiogenesis. A mutation in R-spondin1 (rspo1), a Wnt signaling regulator, was uncovered during a forward-genetic screen for angiogenesis-deficient mutants in the zebrafish. Embryos lacking rspo1 or the proposed rspo1 receptor kremen form primary vessels by vasculogenesis, but are defective in subsequent angiogenesis. Endothelial cell-autonomous inhibition of canonical Wnt signaling also blocks angiogenesis in vivo. The pro-angiogenic effects of Rspo1/Wnt signaling are mediated by Vegfc/Vegfr3(Flt4) signaling. Vegfc expression is dependent on Rspo1 and Wnt, and Vegfc and Vegfr3 are necessary to promote angiogenesis downstream from Rspo1-Wnt. As all of these molecules are expressed by the endothelium during sprouting stages, these results suggest that Rspo1-Wnt-VegfC-Vegfr3 signaling plays a crucial role as an endothelial-autonomous permissive cue for developmental angiogenesis.


Asunto(s)
Neovascularización Fisiológica/genética , Factor C de Crecimiento Endotelial Vascular/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/fisiología , Vía de Señalización Wnt/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Células Cultivadas , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Modelos Biológicos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Trombospondinas , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/genética , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
16.
Dev Dyn ; 242(8): 976-88, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23649798

RESUMEN

BACKGROUND: The major arteries and veins are formed early during development. The molecular tools to identify arterial and venous endothelial cells improve our understanding of arterial-venous differentiation and branching morphogenesis. Compared with arterial differentiation, relatively little is known about what controls venous development, due to lack of definitive molecular markers for venous endothelial cells. RESULTS: Here we report that the antibody against EphB1, an EphB class receptor, makes it possible to establish a reliable whole-mount immunohistochemical analysis of venous identity with greater resolution than previously possible in embryonic and adult skin vasculature models. EphB1 expression is restricted to the entire venous vasculature throughout embryonic development to adulthood, whereas the previously established venous marker EphB4 is also detectable in lymphatic vasculature. This venous-restricted expression of EphB1 is established after the vascular remodeling of the primary capillary plexus has occurred. Compared with its venous-specific expression in the skin, however, EphB1 is not restricted to the venous vasculature in yolk sac, trunk and lung. CONCLUSIONS: These studies introduce EphB1 as a new venous-restricted marker in a tissue-specific and time-dependent manner.


Asunto(s)
Receptor EphB1/metabolismo , Piel/irrigación sanguínea , Animales , Arterias/metabolismo , Células Endoteliales/metabolismo , Inmunohistoquímica , Vasos Linfáticos/metabolismo , Ratones , Receptor EphB4/metabolismo , Venas/metabolismo
17.
Biol Open ; 13(5)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38639409

RESUMEN

Blood vessels serve as intermediate conduits for the extension of sympathetic axons towards target tissues, while also acting as crucial targets for their homeostatic processes encompassing the regulation of temperature, blood pressure, and oxygen availability. How sympathetic axons innervate not only blood vessels but also a wide array of target tissues is not clear. Here we show that in embryonic skin, after the establishment of co-branching between sensory nerves and blood vessels, sympathetic axons invade the skin alongside these sensory nerves and extend their branches towards these blood vessels covered by vascular smooth muscle cells (VSMCs). Our mosaic labeling technique for sympathetic axons shows that collateral branching predominantly mediates the innervation of VSMC-covered blood vessels by sympathetic axons. The expression of nerve growth factor (NGF), previously known to induce collateral axon branching in culture, can be detected in the vascular smooth muscle cell (VSMC)-covered blood vessels, as well as sensory nerves. Indeed, VSMC-specific Ngf knockout leads to a significant decrease of collateral branching of sympathetic axons innervating VSMC-covered blood vessels. These data suggest that VSMC-derived NGF serves as an inductive signal for collateral branching of sympathetic axons innervating blood vessels in the embryonic skin.


Asunto(s)
Músculo Liso Vascular , Factor de Crecimiento Nervioso , Piel , Animales , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/inervación , Factor de Crecimiento Nervioso/metabolismo , Ratones , Piel/inervación , Piel/irrigación sanguínea , Piel/metabolismo , Miocitos del Músculo Liso/metabolismo , Axones/metabolismo , Axones/fisiología , Vasos Sanguíneos/embriología , Vasos Sanguíneos/inervación , Vasos Sanguíneos/metabolismo , Sistema Nervioso Simpático/embriología , Sistema Nervioso Simpático/fisiología , Sistema Nervioso Simpático/metabolismo , Ratones Noqueados
18.
Semin Cell Dev Biol ; 22(9): 1019-27, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21978864

RESUMEN

The nervous system relies on a highly specialized network of blood vessels for development and neuronal survival. Recent evidence suggests that both the central and peripheral nervous systems (CNS and PNS) employ multiple mechanisms to shape the vascular tree to meet its specific metabolic demands, such as promoting nerve-artery alignment in the PNS or the development the blood brain barrier in the CNS. In this article we discuss how the nervous system directly influences blood vessel patterning resulting in neuro-vascular congruence that is maintained throughout development and in the adult.


Asunto(s)
Vasos Sanguíneos/inervación , Encéfalo/irrigación sanguínea , Sistema Nervioso/irrigación sanguínea , Neuronas/fisiología , Animales , Barrera Hematoencefálica , Humanos , Morfogénesis/fisiología
19.
Hum Mol Genet ; 20(16): 3198-206, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21596842

RESUMEN

Cerebral cavernous malformations (CCM) are irregularly shaped and enlarged capillaries in the brain that are prone to hemorrhage, resulting in headaches, seizures, strokes and even death in patients. The disease affects up to 0.5% of the population and the inherited form has been linked to mutations in one of three genetic loci, CCM1, CCM2 and CCM3. To understand the pathophysiology underlying the vascular lesions in CCM, it is critical to develop a reproducible mouse genetic model of this disease. Here, we report that limited conditional ablation of Ccm2 in young adult mice induces observable neurological dysfunction and reproducibly results in brain hemorrhages whose appearance is highly reminiscent of the lesions observed in human CCM patients. We first demonstrate that conventional or endothelial-specific deletion of Ccm2 leads to fatal cardiovascular defects during embryogenesis, including insufficient vascular lumen formation as well as defective arteriogenesis and heart malformation. These findings confirm and extend prior studies. We then demonstrate that the inducible deletion of Ccm2 in adult mice recapitulates the CCM-like brain lesions in humans; the lesions display disrupted vascular lumens, enlarged capillary cavities, loss of proper neuro-vascular associations and an inflammatory reaction. The CCM lesions also exhibit damaged neuronal architecture, the likely cause of neurologic defects, such as ataxia and seizure. These mice represent the first CCM2 animal model for CCM and should provide the means to elucidate disease mechanisms and evaluate therapeutic strategies for human CCM.


Asunto(s)
Envejecimiento/patología , Encéfalo/patología , Eliminación de Gen , Hemangioma Cavernoso del Sistema Nervioso Central/patología , Proteínas de Microfilamentos/metabolismo , Envejecimiento/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Encéfalo/irrigación sanguínea , Modelos Animales de Enfermedad , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Hemangioma Cavernoso del Sistema Nervioso Central/metabolismo , Hemorragia , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Neovascularización Patológica/complicaciones , Neovascularización Patológica/metabolismo
20.
Front Cell Dev Biol ; 11: 1150775, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37091974

RESUMEN

The central nervous system (CNS) is considered as an immune privilege organ, based on experiments in the mid 20th century showing that the brain fails to mount an efficient immune response against an allogeneic graft. This suggests that in addition to the presence of the blood-brain barrier (BBB), the apparent absence of classical lymphatic vasculature in the CNS parenchyma limits the capacity for an immune response. Although this view is partially overturned by the recent discovery of the lymphatic-like hybrid vessels in the Schlemm's canal in the eye and the lymphatic vasculature in the outmost layer of the meninges, the existence of lymphatic vessels in the CNS parenchyma has not been reported. Two potential mechanisms by which lymphatic vasculature may arise in the organs are: 1) sprouting and invasion of lymphatic vessels from the surrounding tissues into the parenchyma and 2) differentiation of blood endothelial cells into lymphatic endothelial cells in the parenchyma. Considering these mechanisms, we here discuss what causes the dearth of lymphatic vessels specifically in the CNS parenchyma.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA