Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
PeerJ ; 9: e11068, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33889441

RESUMEN

BACKGROUND: The fish Totoaba macdonaldi is endemic to the Upper Gulf of California. Its migratory movements involve sites with high levels of trace elements in the environment that can accumulate in tissues. In this study, lead (Pb), copper (Cu), cadmium (Cd), zinc (Zn) and iron (Fe) concentrations in male and female totoaba liver and muscle were quantified at various sexual maturity stages along the species' geographic distribution. METHODS: Generalized linear models were used to explore associations between trace element concentrations and season of the year, sex/maturity stage, and total fish length. RESULTS: No detectable Pb concentrations were recorded in liver or muscle; Cu, Cd, Zn and Fe contents in totoaba liver and muscle were typical of fish inhabiting areas with no contamination issues and are within international maximum permissible levels for human consumption. Variations in the content of Cd, Cu, Zn and Fe in liver of totoaba seem to be more related to the feeding and reproductive physiology of this species than as result of environmental exposure. Results suggest that consumption of totoaba muscle does not pose a public health risk. Furthermore, depending on the sex/maturity stage of totoaba, this fish's muscle may provide approximately 70% Cu, 60% Zn and 100% Fe of the recommended dietary reference intake.

2.
J Cell Biol ; 210(7): 1117-31, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26391660

RESUMEN

Protease-activated receptor 1 (PAR1) is a G protein-coupled receptor (GPCR) for thrombin and promotes inflammatory responses through multiple pathways including p38 mitogen-activated protein kinase signaling. The mechanisms that govern PAR1-induced p38 activation remain unclear. Here, we define an atypical ubiquitin-dependent pathway for p38 activation used by PAR1 that regulates endothelial barrier permeability. Activated PAR1 K63-linked ubiquitination is mediated by the NEDD4-2 E3 ubiquitin ligase and initiated recruitment of transforming growth factor-ß-activated protein kinase-1 binding protein-2 (TAB2). The ubiquitin-binding domain of TAB2 was essential for recruitment to PAR1-containing endosomes. TAB2 associated with TAB1, which induced p38 activation independent of MKK3 and MKK6. The P2Y1 purinergic GPCR also stimulated p38 activation via NEDD4-2-mediated ubiquitination and TAB1-TAB2. TAB1-TAB2-dependent p38 activation was critical for PAR1-promoted endothelial barrier permeability in vitro, and p38 signaling was required for PAR1-induced vascular leakage in vivo. These studies define an atypical ubiquitin-mediated signaling pathway used by a subset of GPCRs that regulates endosomal p38 signaling and endothelial barrier disruption.


Asunto(s)
Endosomas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Receptor PAR-1/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Permeabilidad Capilar/fisiología , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Endosomas/genética , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Células HeLa , Humanos , Ratones , Ratones Noqueados , Ubiquitina-Proteína Ligasas Nedd4 , Receptor PAR-1/genética , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética
3.
Cell Host Microbe ; 14(3): 294-305, 2013 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-24034615

RESUMEN

Cholera toxin (CT), a virulence factor elaborated by Vibrio cholerae, is sufficient to induce the severe diarrhea characteristic of cholera. The enzymatic moiety of CT (CtxA) increases cAMP synthesis in intestinal epithelial cells, leading to chloride ion (Cl(-)) efflux through the CFTR Cl(-) channel. To preserve electroneutrality and osmotic balance, sodium ions and water also flow into the intestinal lumen via a paracellular route. We find that CtxA-driven cAMP increase also inhibits Rab11/exocyst-mediated trafficking of host proteins including E-cadherin and Notch signaling components to cell-cell junctions in Drosophila, human intestinal epithelial cells, and ligated mouse ileal loops, thereby disrupting barrier function. Additionally, CtxA induces junctional damage, weight loss, and dye leakage in the Drosophila gut, contributing to lethality from live V. cholerae infection, all of which can be rescued by Rab11 overexpression. These barrier-disrupting effects of CtxA may act in parallel with Cl(-) secretion to drive the pathophysiology of cholera.


Asunto(s)
Toxina del Cólera/metabolismo , Células Epiteliales/fisiología , Exosomas/efectos de los fármacos , Interacciones Huésped-Patógeno , Proteínas de Uniones Estrechas/antagonistas & inhibidores , Uniones Estrechas/fisiología , Vibrio cholerae/fisiología , Animales , Línea Celular , Cloro/metabolismo , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Drosophila , Células Epiteliales/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Humanos , Ratones , Modelos Biológicos , Sodio/metabolismo , Análisis de Supervivencia , Uniones Estrechas/efectos de los fármacos , Agua/metabolismo
4.
Cancer Res ; 72(22): 5833-42, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22942256

RESUMEN

Kaposi sarcoma, the most common cancer in HIV-positive individuals, is caused by endothelial transformation mediated by the Kaposi sarcoma herpes virus (KSHV)-encoded G-protein-coupled receptor (vGPCR). Infection of blood vascular endothelial cells (BEC) by KSHV reactivates an otherwise silenced embryonic program of lymphatic differentiation. Thus, Kaposi sarcoma tumors express numerous lymphatic endothelial cell (LEC) signature genes. A key unanswered question is how lymphatic reprogramming by the virus promotes tumorigenesis leading to Kaposi sarcoma formation. In this study, we present evidence that this process creates an environment needed to license the oncogenic activity of vGPCR. We found that the G-protein regulator RGS4 is an inhibitor of vGPCR that is expressed in BECs, but not in LECs. RGS4 was downregulated by the master regulator of LEC differentiation PROX1, which is upregulated by KSHV and directs KSHV-induced lymphatic reprogramming. Moreover, we found that KSHV upregulates the nuclear receptor LRH1, which physically interacts with PROX1 and synergizes with it to mediate repression of RGS4 expression. Mechanistic investigations revealed that RGS4 reduced vGPCR-enhanced cell proliferation, migration, VEGF expression, and Akt activation and suppressed tumor formation induced by vGPCR. Our findings resolve long-standing questions about the pathologic impact of KSHV-induced reprogramming of host cell identity, and they offer biologic and mechanistic insights supporting the hypothesis that a lymphatic microenvironment is more favorable for Kaposi sarcoma tumorigenesis.


Asunto(s)
Células Endoteliales/patología , Células Endoteliales/virología , Herpesvirus Humano 8/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Diferenciación Celular/fisiología , Transformación Celular Viral , Regulación hacia Abajo , Células Endoteliales/metabolismo , Femenino , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Proteína Oncogénica v-akt/metabolismo , Regiones Promotoras Genéticas , Proteínas RGS/antagonistas & inhibidores , Proteínas RGS/biosíntesis , Proteínas RGS/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
5.
PLoS Pathog ; 8(6): e1002770, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22719258

RESUMEN

Lymphatic endothelial cells (LECs) are differentiated from blood vascular endothelial cells (BECs) during embryogenesis and this physiological cell fate specification is controlled by PROX1, the master regulator for lymphatic development. When Kaposi sarcoma herpes virus (KSHV) infects host cells, it activates the otherwise silenced embryonic endothelial differentiation program and reprograms their cell fates. Interestingly, previous studies demonstrated that KSHV drives BECs to acquire a partial lymphatic phenotype by upregulating PROX1 (forward reprogramming), but stimulates LECs to regain some BEC-signature genes by downregulating PROX1 (reverse reprogramming). Despite the significance of this KSHV-induced bidirectional cell fate reprogramming in KS pathogenesis, its underlying molecular mechanism remains undefined. Here, we report that IL3 receptor alpha (IL3Rα) and NOTCH play integral roles in the host cell type-specific regulation of PROX1 by KSHV. In BECs, KSHV upregulates IL3Rα and phosphorylates STAT5, which binds and activates the PROX1 promoter. In LECs, however, PROX1 was rather downregulated by KSHV-induced NOTCH signal via HEY1, which binds and represses the PROX1 promoter. Moreover, PROX1 was found to be required to maintain HEY1 expression in LECs, establishing a reciprocal regulation between PROX1 and HEY1. Upon co-activation of IL3Rα and NOTCH, PROX1 was upregulated in BECs, but downregulated in LECs. Together, our study provides the molecular mechanism underlying the cell type-specific endothelial fate reprogramming by KSHV.


Asunto(s)
Células Endoteliales/virología , Infecciones por Herpesviridae/metabolismo , Proteínas de Homeodominio/metabolismo , Receptores de Interleucina-3/metabolismo , Receptores Notch/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula , Células Cultivadas , Ensayo de Cambio de Movilidad Electroforética , Células Endoteliales/metabolismo , Herpesvirus Humano 8/metabolismo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
6.
Circulation ; 125(7): 872-82, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22275501

RESUMEN

BACKGROUND: The lymphatic system plays a key role in tissue fluid homeostasis and lymphatic dysfunction caused by genetic defects, or lymphatic vessel obstruction can cause lymphedema, disfiguring tissue swelling often associated with fibrosis and recurrent infections with no available cures to date. In this study, retinoic acids (RAs) were determined to be a potent therapeutic agent that is immediately applicable to reduce secondary lymphedema. METHODS AND RESULTS: We report that RAs promote proliferation, migration, and tube formation of cultured lymphatic endothelial cells by activating fibroblast growth factor receptor signaling. Moreover, RAs control the expression of cell-cycle checkpoint regulators such as p27(Kip1), p57(Kip2), and the aurora kinases through both an Akt-mediated nongenomic action and a transcription-dependent genomic action that is mediated by Prox1, a master regulator of lymphatic development. Moreover, 9-cisRA was found to activate in vivo lymphangiogenesis in animals in mouse trachea, Matrigel plug, and cornea pocket assays. Finally, we demonstrate that 9-cisRA can provide a strong therapeutic efficacy in ameliorating experimental mouse tail lymphedema by enhancing lymphatic vessel regeneration. CONCLUSION: These in vitro and animal studies demonstrate that 9-cisRA potently activates lymphangiogenesis and promotes lymphatic regeneration in an experimental lymphedema model, presenting it as a promising novel therapeutic agent to treat human lymphedema patients.


Asunto(s)
Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/fisiología , Linfedema/tratamiento farmacológico , Regeneración/efectos de los fármacos , Tretinoina/farmacología , Alitretinoína , Animales , Aurora Quinasas , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Vasos Linfáticos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética , Tretinoina/uso terapéutico
7.
J Ethnopharmacol ; 135(2): 434-9, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21440616

RESUMEN

AIM OF THE STUDY: Ipomoea tyrianthina has been used in Mexican traditional medicine as a mild purgative, for the treatment of nervous disorders, and against tumors. In this study, the effect of convolvulin (an ether-insoluble resin glycoside) from the root of Ipomoea tyrianthina on: Central Nervous System; as spasmolytic and vasodilator; cytotoxic against cancer cell lines is evaluated. MATERIALS AND METHODS: Convolvulin isolated from the root of Ipomoea tyrianthina (IT-EM) was tested on pentylentetrazole induced seizures, pentobarbital-induced hypnosis, release of GABA and glutamic acid, isolated rat aorta and ileum rings, and against Caco-2 and KB cell lines. RESULTS: IT-EM increased the hypnotic effect induced by pentobarbital and the release of GABA in brain cortex of mice, but did not protect mice against pentylenetetrazole-induced convulsions. IT-EM produced a significant vasodilator effect in concentration- and endothelium-dependent manners on isolated rat aorta, but did not inhibit significantly contractions on rat ileum, colon, and jejune rings. IT-EM showed cytotoxic activity against nasopharyngeal carcinoma KB cell line. CONCLUSIONS: Convolvulin (IT-EM) from Ipomoea tyrianthina has sedative effect, vasorelaxant effect in concentration- and endothelium-dependent manners, and cytotoxic activity against nasopharyngeal carcinoma KB cell line.


Asunto(s)
Antineoplásicos/farmacología , Glicósidos/farmacología , Hipnóticos y Sedantes/farmacología , Ipomoea/química , Extractos Vegetales/química , Resinas de Plantas/farmacología , Vasodilatadores/farmacología , Animales , Línea Celular Tumoral , Humanos , Ratones
8.
Blood ; 117(1): 362-5, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20962325

RESUMEN

Although the blood vessel-specific fluorescent transgenic mouse has been an excellent tool to study vasculogenesis and angiogenesis, a lymphatic-specific fluorescent mouse model has not been established to date. Here we report a transgenic animal model that expresses the green fluorescent protein under the promoter of Prox1, a master control gene in lymphatic development. Generated using an approximately 200-kb-long bacterial artificial chromosome harboring the entire Prox1 gene, this Prox1-green fluorescent protein mouse was found to faithfully recapitulate the expression pattern of the Prox1 gene in lymphatic endothelial cells and other Prox1-expressing organs, and enabled us to conveniently visualize detailed structure and morphology of lymphatic vessels and networks throughout development. Our data demonstrate that this novel transgenic mouse can be extremely useful for detection, imaging, and isolation of lymphatic vessels and monitoring wound-associated lymphangiogenesis. Together, this Prox1-green fluorescent protein transgenic mouse will be a great tool for the lymphatic research.


Asunto(s)
Cromosomas Artificiales Bacterianos/genética , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Vasos Linfáticos/citología , Regiones Promotoras Genéticas/genética , Proteínas Supresoras de Tumor/genética , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas de Homeodominio/metabolismo , Humanos , Linfangiogénesis , Vasos Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Supresoras de Tumor/metabolismo
9.
PLoS Pathog ; 6(8): e1001046, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20730087

RESUMEN

Kaposi's sarcoma (KS) is the most common cancer among HIV-positive patients. Histogenetic origin of KS has long been elusive due to a mixed expression of both blood and lymphatic endothelial markers in KS tumor cells. However, we and others discovered that Kaposi's sarcoma herpes virus (KSHV) induces lymphatic reprogramming of blood vascular endothelial cells by upregulating PROX1, which functions as the master regulator for lymphatic endothelial differentiation. Here, we demonstrate that the KSHV latent gene kaposin-B enhances the PROX1 mRNA stability and plays an important role in KSHV-mediated PROX1 upregulation. We found that PROX1 mRNA contains a canonical AU-rich element (ARE) in its 3'-untranslated region that promotes PROX1 mRNA turnover and that kaposin-B stimulates cytoplasmic accumulation of the ARE-binding protein HuR through activation of the p38/MK2 pathway. Moreover, HuR binds to and stabilizes PROX1 mRNA through its ARE and is necessary for KSHV-mediated PROX1 mRNA stabilization. Together, our study demonstrates that kaposin-B plays a key role in PROX1 upregulation during lymphatic reprogramming of blood vascular endothelial cells by KSHV.


Asunto(s)
Células Endoteliales/citología , Regulación de la Expresión Génica , Infecciones por Herpesviridae/metabolismo , Proteínas de Homeodominio/genética , Proteínas Supresoras de Tumor/genética , Proteínas Virales/metabolismo , Regiones no Traducidas 3' , Antígenos de Superficie/metabolismo , Northern Blotting , Western Blotting , Diferenciación Celular/genética , Línea Celular , Proteínas ELAV , Proteína 1 Similar a ELAV , Ensayo de Cambio de Movilidad Electroforética , Células Endoteliales/virología , Infecciones por Herpesviridae/genética , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Proteínas de Homeodominio/biosíntesis , Humanos , Inmunoprecipitación , Estabilidad del ARN/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Regulación hacia Arriba
10.
Blood ; 116(1): 140-50, 2010 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-20351309

RESUMEN

Arteriovenous-lymphatic endothelial cell fates are specified by the master regulators, namely, Notch, COUP-TFII, and Prox1. Whereas Notch is expressed in the arteries and COUP-TFII in the veins, the lymphatics express all 3 cell fate regulators. Previous studies show that lymphatic endothelial cell (LEC) fate is highly plastic and reversible, raising a new concept that all 3 endothelial cell fates may co-reside in LECs and a subtle alteration can result in a reprogramming of LEC fate. We provide a molecular basis verifying this concept by identifying a cross-control mechanism among these cell fate regulators. We found that Notch signal down-regulates Prox1 and COUP-TFII through Hey1 and Hey2 and that activated Notch receptor suppresses the lymphatic phenotypes and induces the arterial cell fate. On the contrary, Prox1 and COUP-TFII attenuate vascular endothelial growth factor signaling, known to induce Notch, by repressing vascular endothelial growth factor receptor-2 and neuropilin-1. We show that previously reported podoplanin-based LEC heterogeneity is associated with differential expression of Notch1 in human cutaneous lymphatics. We propose that the expression of the 3 cell fate regulators is controlled by an exquisite feedback mechanism working in LECs and that LEC fate is a consequence of the Prox1-directed lymphatic equilibrium among the cell fate regulators.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Células Endoteliales/metabolismo , Proteínas de Homeodominio/metabolismo , Receptor Notch1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Western Blotting , Factor de Transcripción COUP II/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Células Cultivadas , Regulación hacia Abajo , Células Endoteliales/citología , Retroalimentación Fisiológica , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Interferencia de ARN , Receptor Notch1/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Serrate-Jagged , Transducción de Señal , Proteínas Supresoras de Tumor/genética
11.
Anal Biochem ; 386(2): 251-5, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19150324

RESUMEN

Although various nonviral transfection methods are available, cell toxicity, low transfection efficiency, and high cost remain hurdles for in vitro gene delivery in cultured primary endothelial cells. Recently, unprecedented transfection efficiency for primary endothelial cells has been achieved due to the newly developed nucleofection technology that uses a combination of novel electroporation condition and specific buffer components that stabilize the cells in the electrical field. Despite superior transfection efficiency and cell viability, high cost of the technology has discouraged cardiovascular researchers from liberally adopting this new technology. Here we report that a phosphate-buffered saline (PBS)-based nucleofection method can be used for efficient gene delivery into primary endothelial cells and other types of cells. Comparative analyses of transfection efficiency and cell viability for primary arterial, venous, microvascular, and lymphatic endothelial cells were performed using PBS. Compared with the commercial buffers, PBS can support equally remarkable nucleofection efficiency to both primary and nonprimary cells. Moreover, PBS-mediated nucleofection of small interfering RNA (siRNA) showed more than 90% knockdown of the expression of target genes in primary endothelial cells. We demonstrate that PBS can be an unprecedented economical alternative to the high-cost buffers or nucleofection of various primary and nonprimary cells.


Asunto(s)
Electroporación/métodos , Células Endoteliales/metabolismo , Cloruro de Sodio/química , Transfección/métodos , Tampones (Química) , Supervivencia Celular , Humanos , Fosfatos/química , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
12.
Blood ; 113(8): 1856-9, 2009 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-18815287

RESUMEN

Specification of endothelial cell (EC) fate during vascular development is controlled by distinct key regulators. While Notch plays an essential role in induction of arterial phenotypes, COUP-TFII is required to maintain the venous EC identity. Homeodomain transcription factor Prox1 functions to reprogram venous ECs to lymphatic endothelial cells (LECs). Here, we report that the venous EC fate regulator COUP-TFII is expressed in LECs throughout development and physically interacts with Prox1 to form a stable complex in various cell types including LECs. We found that COUP-TFII functions as a coregulator of Prox1 to control several lineage-specific genes including VEGFR-3, FGFR-3, and neuropilin-1 and is required along with Prox1 to maintain LEC phenotype. Together, we propose that the physical and functional interactions of the 2 proteins constitute an essential part in the program specifying LEC fate and may provide the molecular basis for the hypothesis of venous EC identity being the prerequisite for LEC specification.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Células Endoteliales/citología , Células Endoteliales/fisiología , Proteínas de Homeodominio/metabolismo , Sistema Linfático/embriología , Proteínas Supresoras de Tumor/metabolismo , Factor de Transcripción COUP II/genética , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Sistema Linfático/citología , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA