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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arch Toxicol ; 92(5): 1877-1891, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29556671

RESUMEN

A high incidence of hemangiosarcoma (HSA) was observed in mice treated for 2 years with siponimod, a sphingosine-1-phosphate receptor 1 (S1P1) functional antagonist, while no such tumors were observed in rats under the same treatment conditions. In 3-month rat (90 mg/kg/day) and 9-month mouse (25 and 75 mg/kg/day) in vivo mechanistic studies, vascular endothelial cell (VEC) activation was observed in both species, but VEC proliferation and persistent increases in circulating placental growth factor 2 (PLGF2) were only seen in the mouse. In mice, these effects were sustained over the 9-month study duration, while in rats increased mitotic gene expression was present at day 3 only and PLGF2 was induced only during the first week of treatment. In the mouse, the persistent VEC activation, mitosis induction, and PLGF2 stimulation likely led to sustained neo-angiogenesis which over life-long treatment may result in HSA formation. In rats, despite sustained VEC activation, the transient mitotic and PLGF2 stimuli did not result in the formation of HSA. In vitro, the mouse and rat primary endothelial cell cultures mirrored their respective in vivo findings for cell proliferation and PLGF2 release. Human VECs, like rat cells, were unresponsive to siponimod treatment with no proliferative response and no release of PLGF2 at all tested concentrations. Hence, it is suggested that the human cells also reproduce a lack of in vivo response to siponimod. In conclusion, the molecular mechanisms leading to siponimod-induced HSA in mice are considered species specific and likely irrelevant to humans.


Asunto(s)
Azetidinas/efectos adversos , Compuestos de Bencilo/efectos adversos , Células Endoteliales/efectos de los fármacos , Hemangiosarcoma/inducido químicamente , Pruebas de Toxicidad Crónica/métodos , Administración Oral , Animales , Azetidinas/administración & dosificación , Compuestos de Bencilo/administración & dosificación , Células Cultivadas , Endotelio Vascular/citología , Hemangiosarcoma/genética , Humanos , Masculino , Ratones Endogámicos , Factor de Crecimiento Placentario/metabolismo , Ratas Sprague-Dawley , Ratas Wistar , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/metabolismo , Especificidad de la Especie , Toxicocinética , Transcriptoma/efectos de los fármacos
2.
Int J Toxicol ; 30(3): 300-12, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21653914

RESUMEN

This article addresses the issue of miscorrelation between hepatic injury biomarkers and histopathological findings in the drug development context. Our studies indicate that the use of toxicogenomics can aid in the drug development decision-making process associated with such miscorrelated data. BLZ945 was developed as a Colony-Stimulating Factor 1 Receptor (CSF-1R) inhibitor. Treatment of BLZ945 in rats and monkeys increased serum alanine aminotransferase (ALT) and aspartate transaminase (AST). However, liver hypertrophy was the only histopathological liver finding in rats, and there was no change in the livers of monkeys. Longer treatment of BLZ945 in rats for 6 weeks caused up to 6-fold elevation of ALT, yet hepatocyte necrosis was not detected microscopically. Toxicogenomic profiling of liver samples demonstrated that the genes associated with early response to liver injury, apoptosis/necrosis, inflammation, oxidative stress, and metabolic enzymes were upregulated. Studies are ongoing to evaluate the mechanisms underlying BL945-induced ALT and AST elevations.


Asunto(s)
Benzotiazoles/toxicidad , Biomarcadores/sangre , Hígado/efectos de los fármacos , Farmacogenética , Ácidos Picolínicos/toxicidad , Pruebas de Toxicidad , Alanina Transaminasa/sangre , Alanina Transaminasa/genética , Animales , Aspartato Aminotransferasas/sangre , Aspartato Aminotransferasas/genética , Regulación hacia Abajo/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Hígado/enzimología , Hígado/patología , Macaca fascicularis , ARN Mensajero/genética , Ratas , Ratas Wistar , Regulación hacia Arriba/efectos de los fármacos
3.
Novartis Found Symp ; 283: 220-7; discussion 227-9, 238-41, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18300425

RESUMEN

In the human, malformations of lymphatic vessels can be observed as lymphangiectasia, lymphangioma and lymphangiomatosis, with a prevalence of 1.2-2.8 per thousand. Their aetiology is unknown and a causal therapy does not exist. We investigated the origin of lymphatic endothelial cells (LECs) in avian and murine embryos, and compared the molecular profile of LECs from normal and malformed lymphatics of children. In avian embryos, Prox1+ lymphangioblasts are located in the confluence of the cranial and caudal cardinal veins, where the jugular lymph sac (JLS) forms. Cell lineage studies show that the JLS is of venous origin. In contrast, the lymphatics of the dermis are derived from mesenchymal lymphangioblasts located in the dermatomes, suggesting a dual origin of LECs in avian embryos. The same may hold true for murine embryos, where Lyve1+ LEC precursors are found in the cardinal veins, and in the mesenchyme. The mesenchymal cells express the pan-leukocyte marker CD45, indicating a cell type with lymphendothelial and leukocyte characteristics. In the human, such cells might give rise to Kaposi's sarcoma. Microarray analyses of LECs from lymphangiomas of children show a large number of regulated genes, such as VEGFR3. Our studies show that lymphvasculogenesis and lymphangiogenesis occur simultaneously in the embryo, and suggest a function for VEGFR3 in lymphangiomas.


Asunto(s)
Desarrollo Embrionario , Vasos Linfáticos/anomalías , Vasos Linfáticos/embriología , Animales , Línea Celular , Embrión de Pollo , Niño , Embrión no Mamífero/citología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Hibridación in Situ , Ratones , Codorniz , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
4.
BMC Cancer ; 7: 105, 2007 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-17584927

RESUMEN

BACKGROUND: Lymphangiomas are neoplasias of childhood. Their etiology is unknown and a causal therapy does not exist. The recent discovery of highly specific markers for lymphatic endothelial cells (LECs) has permitted their isolation and characterization, but expression levels and stability of molecular markers on LECs from healthy and lymphangioma tissues have not been studied yet. We addressed this problem by profiling LECs from normal dermis and two children suffering from lymphangioma, and also compared them with blood endothelial cells (BECs) from umbilical vein, aorta and myometrial microvessels. METHODS: Lymphangioma tissue samples were obtained from two young patients suffering from lymphangioma in the axillary and upper arm region. Initially isolated with anti-CD31 (PECAM-1) antibodies, the cells were separated by FACS sorting and magnetic beads using anti-podoplanin and/or LYVE-1 antibodies. Characterization was performed by FACS analysis, immunofluorescence staining, ELISA and micro-array gene analysis. RESULTS: LECs from foreskin and lymphangioma had an almost identical pattern of lymphendothelial markers such as podoplanin, Prox1, reelin, cMaf and integrin-alpha1 and -alpha9. However, LYVE-1 was down-regulated and VEGFR-2 and R-3 were up-regulated in lymphangiomas. Prox1 was constantly expressed in LECs but not in any of the BECs. CONCLUSION: LECs from different sources express slightly variable molecular markers, but can always be distinguished from BECs by their Prox1 expression. High levels of VEGFR-3 and -2 seem to contribute to the etiology of lymphangiomas.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Linfangioma/metabolismo , Neoplasias Cutáneas/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adolescente , Biopsia con Aguja , Estudios de Casos y Controles , Niño , Células Endoteliales/citología , Endotelio Linfático/citología , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Linfangioma/patología , Masculino , Pronóstico , Proteína Reelina , Valores de Referencia , Muestreo , Sensibilidad y Especificidad , Neoplasias Cutáneas/patología , Células Tumorales Cultivadas
5.
Oncogene ; 24(4): 680-7, 2005 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-15580313

RESUMEN

Amplification of the MYCN oncogene contributes to the malignant progression of human neuroblastomas, but the mechanisms have remained unclear. We have previously demonstrated that N-Myc facilitates angiogenesis by downregulating an angiogenesis inhibitor identified as the inhibin betaA homodimer activin A. Here, we have sought to define the molecular, biological and clinical consequences of activin A expression in human neuroblastoma. We report that enhanced activin A expression suppresses proliferation and colony formation of human neuroblastoma cells with amplified MYCN in vitro; that it inhibits neuroblastoma growth and angiogenesis in vivo; that it is highly expressed in differentiated, but not undifferentiated human neuroblastomas; and that it correlates with favourable outcome of neuroblastoma patients. Our results indicate that high activin A expression plays an important beneficial role in human neuroblastoma.


Asunto(s)
Activinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Subunidades beta de Inhibinas/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Activinas/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia sin Enfermedad , Humanos , Inmunohistoquímica , Subunidades beta de Inhibinas/genética , Ratones , Trasplante de Neoplasias , Neuroblastoma/diagnóstico , Neuroblastoma/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Resultado del Tratamiento
6.
Cancer Res ; 64(17): 6109-18, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15342394

RESUMEN

Neuroblastoma is the most frequent solid childhood malignancy. Despite aggressive therapy, mortality is high due to rapid tumor progression to advanced stages. The molecules and mechanisms underlying poor prognosis are not well understood. Here, we report that cultured human neuroblastoma cells express the hepatocyte growth factor (HGF) and its receptor c-Met. Binding of HGF to c-Met triggers receptor autophosphorylation, indicating functional relevance of this interaction. HGF activates several downstream effectors of c-Met such as the mitogen-activated protein kinases extracellular signal-regulated kinase 1/extracellular signal-regulated kinase 2 and phospholipase C-gamma, whereas signal transducer and activator of transcription 3 is constitutively activated in neuroblastoma cells expressing c-Met. In addition, HGF is able to stimulate expression and proteolytic activity of matrix metalloproteinase-2 and tissue-type plasminogen activator in neuroblastoma cells, thereby promoting degradation of extracellular matrix components. We show that HGF stimulates invasion of neuroblastoma cells in vitro and in vivo, and it promotes the formation of angiogenic neuroblastomas in vivo. These processes can be blocked by specific inhibitors of the mitogen-activated protein kinase cascade, by inhibitors of phospholipase C-gamma, and also by the expression of a dominant negative signal transducer and activator of transcription 3 mutant. Our data provide the first evidence that the HGF/c-Met pathway is essential for invasiveness and malignant progression of human neuroblastomas. They further suggest that specific inhibitors of this pathway may be suitable as therapeutic agents to improve clinical outcome of neuroblastomas.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , División Celular/fisiología , Línea Celular Tumoral , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Factor de Crecimiento de Hepatocito/biosíntesis , Humanos , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 2 de la Matriz/metabolismo , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neuroblastoma/irrigación sanguínea , Neuroblastoma/genética , Fosforilación , Proteínas Proto-Oncogénicas c-met/biosíntesis , Factor de Transcripción STAT3 , Activador de Tejido Plasminógeno/metabolismo , Transactivadores/biosíntesis , Transactivadores/genética , Transactivadores/metabolismo , Transfección
7.
FASEB J ; 16(10): 1271-3, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12060670

RESUMEN

Detection of lymphatic endothelal cells (LECs) has been problematic because of the lack of specific markers. The homeobox transcription factor Prox1 is expressed in LECs of murine and avian embryos. We have studied expression of Prox1 in human tissues with immunofluorescence. In 19-wk-old human fetuses, Prox1 and vascular endothelial growth factor receptor-3 (VEGFR-3) are coexpressed in LECs of lymphatic trunks and lymphatic capillaries. Prox1 is located in the nucleus, and its expression is mutually exclusive with that of the blood vascular marker PAL-E. Prox1 is a constitutive marker of LECs and is found in tissues of healthy adults and lymphedema patients. Blood vascular endothelial cells (BECs) of hemangiomas express CD31 and CD34, but not Prox1. A subset of these cells is positive for VEGFR-3. Lymphatics in the periphery of hemangiomas express Prox1 and CD31, but not CD34. In lymphangiomas, LECs express Prox1, CD31, and VEGFR-3, but rarely CD34. In the stroma, spindle-shaped CD34-positive cells are present. We show that Prox1 is a reliable marker for LECs in normal and pathologic human tissues, coexpressed with VEGFR-3 and CD31. VEGFR-3 and CD34 are less reliable markers for LECs and BECs, respectively, because exceptions from their normal expression patterns are found in pathologic tissues.


Asunto(s)
Endotelio Linfático/química , Proteínas de Homeodominio/análisis , Enfermedades Linfáticas/metabolismo , Factores de Transcripción/análisis , Antígenos CD34/análisis , Biomarcadores/análisis , Biomarcadores de Tumor/análisis , Preescolar , Endotelio Linfático/metabolismo , Hemangioma/química , Proteínas de Homeodominio/metabolismo , Humanos , Lactante , Recién Nacido , Linfangioma/química , Linfedema/metabolismo , Especificidad de Órganos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor , Receptor 3 de Factores de Crecimiento Endotelial Vascular
8.
Anat Embryol (Berl) ; 207(1): 29-34, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12768422

RESUMEN

Cells from the ventrolateral dermomyotomal lips at limb levels undergo epithelio-mesenchymal transition and migrate as individual and undifferentiated cells into the limb buds. The cells give rise to myocytes and blood vascular endothelial cells (BECs) in the limb. Using vascular endothelial growth factor receptor-3 (VEGFR-3) as a marker, it has also been shown that the somites contribute to endothelial cells of lymphatic vessels in the limbs, but it is unknown where the lymphangiogenic precursors are located within the somite. In this study we used the transcription factor Prox1 as a lymphatic marker and investigated whether cells in the dorso-lateral quarter of the somite differentiate into lymphatic endothelial cells (LECs) of the limbs. To label the migrating cells, the dorso-lateral part of an epithelial brachial somite was grafted homotopically from quail into chick embryos at HH stages 13-14. The chick hosts were incubated until day 10-11 of development. The quail cell nuclei were identified with QCPN (anti-quail) antibodies. Cell differentiation was analysed by immunohistochemical staining with QH1, anti-desmin and anti-Prox1 antibodies, and by in situ hybridisation with Prox1 probes. Our results confirm that quail cell nuclei are incorporated into the myotubes of the limb muscles. Quail cells are found in the endothelium of limb blood vessels and lymphatics, predominantly the dermal lymphatics. This indicates that superficial lymphatics develop independently from the deep ones and shows that cells migrating from the lateral somitic edge into the limb buds differentiate into three cell populations: myocytes, BECs and LECs.


Asunto(s)
Movimiento Celular/fisiología , Esbozos de los Miembros/embriología , Vasos Linfáticos/embriología , Somitos/metabolismo , Células Madre/metabolismo , Animales , Biomarcadores , Vasos Sanguíneos/citología , Vasos Sanguíneos/embriología , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Embrión de Pollo , Dermis/citología , Dermis/embriología , Desmina/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Extremidades/embriología , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Esbozos de los Miembros/citología , Esbozos de los Miembros/metabolismo , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Codorniz , Somitos/citología , Células Madre/citología , Trasplante Heterólogo , Proteínas Supresoras de Tumor
10.
Dev Biol ; 305(2): 451-9, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17383624

RESUMEN

The mass of the myocardium and endocardium of the vertebrate heart derive from the heart-forming fields of the lateral plate mesoderm. Further components of the mature heart such as the epicardium, cardiac interstitium and coronary blood vessels originate from a primarily extracardiac progenitor cell population: the proepicardium (PE). The coronary blood vessels are accompanied by lymph vessels, suggesting a common origin of the two vessel types. However, the origin of cardiac lymphatics has not been studied yet. We have grafted PE of HH-stage 17 (day 3) quail embryos hetero- and homotopically into chick embryos, which were re-incubated until day 15. Double staining with the quail endothelial cell (EC) marker QH1 and the lymphendothelial marker Prox1 shows that the PE of avian embryos delivers hemangioblasts but not lymphangioblasts. We have never observed quail ECs in lymphatics of the chick host. However, one exception was a large lymphatic trunk at the base of the chick heart, indicating a lympho-venous anastomosis and a 'homing' mechanism of venous ECs into the lymphatic trunk. Cardiac lymphatics grow from the base toward the apex of the heart. In murine embryos, we observed a basal to apical gradient of scattered Lyve-1+/CD31+/CD45+ cells in the subepicardium at embryonic day 12.5, indicating a contribution of immigrating lymphangioblasts to the cardiac lymphatic system. Our studies show that coronary blood and lymph vessels are derived from different sources, but grow in close association with each other.


Asunto(s)
Movimiento Celular/fisiología , Vasos Coronarios/embriología , Endotelio Linfático/embriología , Endotelio Vascular/embriología , Células Madre Hematopoyéticas/citología , Vasos Linfáticos/embriología , Pericardio/citología , Pericardio/embriología , Animales , Embrión de Pollo , Vasos Coronarios/citología , Coturnix/embriología , Endotelio Linfático/citología , Endotelio Vascular/citología , Células Madre Hematopoyéticas/fisiología , Linfangiogénesis/fisiología , Vasos Linfáticos/citología , Ratones
11.
Cell Tissue Res ; 330(2): 209-20, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17828556

RESUMEN

The homeobox transcription factor Prox1 is expressed in embryonic hepatoblasts and remains expressed in adult hepatocytes. Prox1-null mice show severe deficiencies in liver development, although the underlying mechanisms are unknown. We have studied the effects of Prox1 on the transcriptional profile of met-murine hepatocytes (MMH) obtained on embryonic day 14 (ED14). These immortalized murine hepatoblasts express numerous hepatoblast markers, but not Prox1. We have performed stable transfection with Prox1 cDNA, analyzed the transcriptome with Agilent mouse whole-genome microarrays, and validated genes by quantitative reverse transcription/polymerase chain reaction. We have observed the up-regulation of 22 genes and the down-regulation of 232 genes, by more than 12-fold. Many of these genes are involved in metabolic hepatocyte functions and may be regulated by Prox1 directly or indirectly, e.g., by the down-regulation of hepatocyte nuclear factor 4alpha. Prox1 induces the down-regulation of transcription factors that are highly expressed in neighboring endodermal organs, suggesting a function during hepatoblast commitment. Prox1 does not influence the proliferative activity of MMH but regulates genes involved in liver morphogenesis. We have observed the up-regulation of both type-IValpha3 procollagen and functionally active matrix metalloproteinase-2 (MMP-2), an observation that places Prox1 at the center of liver matrix turnover. This is consistent with MMP-2 expression in hepatoblasts during liver development and with the persistence of a basal lamina around the liver bud in Prox1-deficient mice. Our studies suggest that Prox1 is a multifunctional regulator of liver morphogenesis and of hepatocyte function and commitment.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Hepatocitos/metabolismo , Proteínas de Homeodominio/metabolismo , Hígado/embriología , Hígado/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Células Madre/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , Linaje de la Célula/genética , Células Cultivadas , Colágeno Tipo IV/metabolismo , Regulación hacia Abajo/genética , Proteínas de Homeodominio/genética , Humanos , Hígado/citología , Ratones , Ratas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Proteínas Supresoras de Tumor/genética
12.
Dev Dyn ; 236(10): 2952-61, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17879316

RESUMEN

Lymphangioma is a disfiguring malformation of early childhood. A mouse lymphangioma model has been established by injecting Freund's incomplete adjuvant (FIA) intraperitoneally, but has not been compared with the human disease. We show that, in accordance with studies from the 1960s, the mouse model represents an oil-granuloma, made up of CD45-positive leukocytes and invaded by blood and lymph vessels. Several markers of lymphatic endothelial cells are expressed in both mouse and human, like CD31, Prox1, podoplanin, and Lyve-1. However, the human disease affects all parts of the lymphovascular tree. We observed convolutes of lymphatic capillaries, irregularly formed collectors with signs of disintegration, and large lymph cysts. We observed VEGFR-2 and -3 expression in both blood vessels and lymphatics of the patients, whereas in mouse VEGFR-2 was confined to activated blood vessels. The experimental mouse FIA model represents a vascularized oil-granuloma rather than a lymphangioma and reflects the complexity of human lymphangioma only partially.


Asunto(s)
Modelos Animales de Enfermedad , Linfangioma , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Animales , Biomarcadores/metabolismo , Endotelio Linfático/patología , Adyuvante de Freund , Granuloma/metabolismo , Granuloma/patología , Humanos , Lactante , Linfangioma/irrigación sanguínea , Linfangioma/metabolismo , Linfangioma/patología , Vasos Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Dev Dyn ; 235(6): 1554-62, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16502417

RESUMEN

The development of lymphatic endothelial cells (LECs) from deep embryonic veins or mesenchymal lymphangioblasts is controversially discussed. Studies employing quail-chick grafting experiments have shown that various mesodermal compartments of the embryo possess lymphangiogenic potential, whereas studies on murine embryos have been in favor of a venous origin of LECs. We have investigated NMRI mice from embryonic day (ED) 9.5 to 13.5 with antibodies against the leukocyte marker CD45, the pan-endothelial marker CD31, and the lymphendothelial markers Prox1 and Lyve-1. Early signs of the development of lymphatics are the Lyve-1- and Prox1-positive segments of the jugular and vitelline veins. Then, lymph sacs, which are found in the jugular region of ED 11.5 mice, express Prox1, Lyve-1, and CD31. Furthermore, scattered cells positive for all of the four markers are present in the mesenchyme of the dermatomes and the mediastinum before lymphatic vessels are present in these regions. Their number increases during development. A gradient of increasing CD31 expression can be seen the closer the cells are located to the lymph sacs. Our studies provide evidence for the existence of scattered mesenchymal cells, which up-regulate lymphendothelial and down-regulate leukocyte characteristics when they integrate into growing murine lymphatics. Such stem cells may also be present in the human and may be the cell of origin in post-transplantation Kaposi sarcoma.


Asunto(s)
Células Endoteliales/fisiología , Leucocitos/fisiología , Mesodermo/citología , Células Madre/fisiología , Animales , Linfangiogénesis/fisiología , Ratones
14.
Dev Biol ; 292(1): 165-73, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16457798

RESUMEN

The earliest signs of the lymphatic vascular system are the lymph sacs, which develop adjacent to specific embryonic veins. It has been suggested that sprouts from the lymph sacs form the complete lymphatic vascular system. We have studied the origin of the jugular lymph sacs (JLS), the dermal lymphatics and the lymph hearts of avian embryos. In day 6.5 embryos, the JLS is an endothelial-lined sinusoidal structure. The lymphatic endothelial cells (LECs) stain (in the quail) positive for QH1 antibody and soybean agglutinin. As early as day 4, the anlagen of the JLS can be recognized by their Prox1 expression. Prox1 is found in the jugular section of the cardinal veins, and in scattered cells located in the dermatomes along the cranio-caudal axis and in the splanchnopleura. In the quail, such cells are positive for Prox1 and QH1. In the jugular region, the veins co-express the angiopoietin receptor Tie2. Quail-chick-chimera studies show that the peripheral parts of the JLS form by integration of cells from the paraxial mesoderm. Intra-venous application of DiI-conjugated acetylated low-density lipoprotein into day 4 embryos suggests a venous origin of the deep parts of the JLS. Superficial lymphatics are directly derived from the dermatomes, as shown by dermatome grafting. The lymph hearts in the lumbo-sacral region develop from a plexus of Prox1-positive lymphatic capillaries. Both LECs and muscle cells of the lymph hearts are of somitic origin. In sum, avian lymphatics are of dual origin. The deep parts of the lymph sacs are derived from adjacent veins, the superficial parts of the JLS and the dermal lymphatics from local lymphangioblasts.


Asunto(s)
Embrión de Pollo , Sistema Linfático/embriología , Codorniz/embriología , Animales , Embrión de Pollo/irrigación sanguínea , Embrión de Pollo/citología , Pollos , Técnica del Anticuerpo Fluorescente , Proteínas de Homeodominio/metabolismo , Hibridación in Situ , Sistema Linfático/citología , Sistema Linfático/metabolismo , Codorniz/fisiología , Receptor TIE-2/metabolismo , Proteínas Supresoras de Tumor
15.
Histochem Cell Biol ; 126(5): 549-62, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16770575

RESUMEN

The aim of this study was to analyse the changes of Prospero-related homeobox 1 (Prox1) gene expression in rat liver under different experimental conditions of liver injury, regeneration and acute phase reaction, and to correlate it with that of markers for hepatoblasts, hepatocytes, cholangiocytes and oval cells. Gene expression was studied at RNA level by RT-PCR, and at protein level by immunohistochemistry. At embryonal stage of rat liver development (embryonal days (ED) 14-16) hepatoblasts were found to be Prox1(+)/Cytokeratin (CK) 19(+) and alpha-fetoprotein (AFP)(+), at this stage Prox1(-)/CK19(+)/AFP(-) small cells (early cholangiocytes?) were identified. In fetal liver (ED 18-22) hepatoblasts were Prox1(+)/CK19(-)/AFP(+). CK7(+) cholangiocytes were detected at this stage, and they were Prox1(-)/AFP(-). In the adult liver hepatocytes were Prox1(+)/CK19(-)/CK7(-)/AFP(-), cholangiocytes were CK19(+) and/or CK7(+) and AFP(-)/Prox1(-). In models of liver damage and regeneration Prox1 remained a stable marker of hepatocytes. After 2-acetyl-aminofluorene treatment with partial hepatectomy (AAF/PH) the amount of Prox1 specific transcripts was low in the liver, when CK19 and AFP gene expression was high, and at no time point AFP(+)/CK19(+ )"oval cells" were found to be Prox1(+). However, a few Prox1(+)/CK19(+) and a few Prox1(+)/CK7(+ )cells were identified in the liver of AAF/PH-animals, which may represent precursors of hepatocytes, or a precancerous state.


Asunto(s)
Hepatocitos/metabolismo , Proteínas de Homeodominio/metabolismo , Regeneración Hepática/fisiología , Hígado/embriología , Proteínas Supresoras de Tumor/metabolismo , 2-Acetilaminofluoreno , Reacción de Fase Aguda/metabolismo , Animales , Conductos Biliares/metabolismo , Biomarcadores/metabolismo , Intoxicación por Tetracloruro de Carbono/patología , Enfermedad Hepática Inducida por Sustancias y Drogas , Células Epiteliales/metabolismo , Femenino , Inmunohistoquímica , Queratina-19/biosíntesis , Queratina-7/biosíntesis , Hígado/patología , Hepatopatías/metabolismo , Masculino , Embarazo , Ratas , Ratas Endogámicas F344 , Ratas Wistar , alfa-Fetoproteínas/biosíntesis
16.
Histochem Cell Biol ; 118(3): 241-9, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12271360

RESUMEN

The microcirculation of tumors is severely disturbed. Tumors are usually supplied by fragile capillaries and do not possess the natural hierarchy of blood vessels. The detection of specific markers for arterial and venous endothelial cells (ECs) now enables us to study the vascular tree in tumors. We have injected rat C6 glioma and human A375 melanoma cells into 3.5- to 4-day-old avian embryos. After 10-12 days of reincubation the tumor cells formed solid tumors vascularized by host ECs. In contrast to the melanomas, the gliomas induced an almost normal vascular tree with arterial and venous vessels. The arterial vessels express the arterial EC marker ephrin-B2, and possess a media of smooth muscle alpha-actin (alphaSMA)-positive cells. Venular vessels in the gliomas are ephrin-B2-negative/alphaSMA-positive. Although the gliomas may represent a rare case of vascular tree induction in tumors, the results underline the heterogeneity of tumor-induced angiogenesis. This has an impact on tumor blood flow and thereby also on the efficacy of chemotherapy and radiotherapy.


Asunto(s)
Glioma/irrigación sanguínea , Melanoma/irrigación sanguínea , Neovascularización Patológica/patología , Actinas/análisis , Animales , Embrión de Pollo , Embrión no Mamífero/irrigación sanguínea , Efrina-B2/análisis , Glioma/patología , Humanos , Inmunohistoquímica , Hibridación in Situ , Melanoma/patología , Músculo Liso/química , Trasplante de Neoplasias , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Codorniz , Ratas , Trasplante Heterólogo , Células Tumorales Cultivadas , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
17.
Histochem Cell Biol ; 117(6): 527-34, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12107503

RESUMEN

The role of pericytes (PCs) during embryonic or tumor angiogenesis is a matter of debate. We studied the expression of cytoskeletal, membrane, and matrix markers in experimental tumors of the human mammary ductal adenoma MDA-MB231 cell line that were grown on avian chorioallantoic membranes (CAMs) from incubation day 10 to 18 (chick) or 8 to 15 (quail). The expression patterns of alpha-smooth muscle actin (alphaSMA) and desmin, of adhesion molecules beta1 integrin and neurothelin, and of fibronectin and laminin were analyzed with conventional and confocal laser scanning microscopy. The CAM arterial wall showed strong alphaSMA signal in all smooth muscle cell layers but the innermost layer, which was desmin positive. Ramified alphaSMA-negative cells with delicate desmin staining accompanied most minor vessels and were also seen basal to the capillary plexus indicating the presence of PCs. In the tumor nodules, a diffuse alphaSMA signal without definite relationship to vascular structures was detected. Strongly desmin-positive, alphaSMA-negative cells were frequent in the zone of contact to the CAM in small nodules, and were scattered in larger tumors. In some regions they were associated with microvessels, and in others appeared detaching from endothelial cells (ECs) or as single migrating cells. We conclude that: (a) the CAM tumor angiogenesis assay is useful for studying PC/EC interactions, (b) PCs are recruited from the CAM into experimental tumor nodules, (c) variability of vasculature in MDA-MB231 tumors may be due to variable PC/EC interactions, and (d) alphaSMA should be used with caution as a general PC marker.


Asunto(s)
Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica/patología , Pericitos/patología , Animales , Comunicación Celular , Membrana Celular , Embrión de Pollo , Citoesqueleto , Endotelio Vascular/patología , Matriz Extracelular , Humanos , Microscopía Fluorescente , Neoplasias Experimentales/patología , Pericitos/ultraestructura , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA