Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Oncogene ; 25(12): 1721-32, 2006 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-16288224

RESUMEN

The highly invasive behavior of glioblastoma cells contributes to the morbidity and mortality associated with these tumors. The integrin-mediated adhesion and migration of glioblastoma cells on brain matrix proteins is enhanced by stimulation with growth factors, including platelet-derived growth factor (PDGF). As focal adhesion kinase (FAK), a nonreceptor cytoplasmic tyrosine kinase, has been shown to promote cell migration in various other cell types, we analysed its role in glioblastoma cell migration. Forced overexpression of FAK in serum-starved glioblastoma cells plated on recombinant (rec)-osteopontin resulted in a twofold enhancement of basal migration and a ninefold enhancement of PDGF-BB-stimulated migration. Both expression of mutant FAK(397F) and the downregulation of FAK with small interfering (si) RNA inhibited basal and PDGF-stimulated migration. FAK overexpression and PDGF stimulation was found to increase the phosphorylation of the Crk-associated substrate (CAS) family member human enhancer of filamentation 1 (HEF1), but not p130CAS or Src-interacting protein (Sin)/Efs, although the levels of expression of these proteins was similar. Moreover downregulation of HEF1 with siRNA, but not p130CAS, inhibited basal and PDGF-stimulated migration. The phosphorylated HEF1 colocalized with vinculin and was associated almost exclusively with 0.1% Triton X-100 insoluble material, consistent with its signaling at focal adhesions. FAK overexpression promoted invasion through normal brain homogenate and siHEF1 inhibited this invasion. Results presented here suggest that HEF1 acts as a necessary and specific downstream effector of FAK in the invasive behavior of glioblastoma cells and may be an effective target for treatment of these tumors.


Asunto(s)
Neoplasias Encefálicas/patología , Movimiento Celular/fisiología , Quinasa 1 de Adhesión Focal/metabolismo , Glioblastoma/patología , Invasividad Neoplásica/patología , Fosfoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Glioblastoma/metabolismo , Humanos , Immunoblotting , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/metabolismo , ARN Interferente Pequeño
2.
J Clin Invest ; 88(6): 1924-32, 1991 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1721625

RESUMEN

Glioblastoma multiforme, the most malignant astroglial-derived tumor, grows as an adherent mass and locally invades normal brain. An examination of adult cerebral glioblastoma biopsy material for the expression of adhesive proteins that might potentiate adhesion and invasion demonstrated tumor cell-associated vitronectin (5/5). In contrast, vitronectin was not detected associated with glial cells in low grade astroglial tumors (0/4), reactive astrogliosis (0/4), or in normal adult cortex and cerebral white matter (0/5). Also, a wide variety of other adhesive ligands were absent from the glioblastoma tumor parenchyma. The alpha v beta 3 integrin was the only vitronectin receptor identified in glioblastoma tumors in situ, and was also not expressed on low grade astroglial-derived tumors, reactive astrogliosis, or on glia or neurons in normal adult cortex and cerebral white matter. In a cell attachment assay, cultured glioblastoma cells attached to the parenchyma of glioblastoma tumor cryostat sections at the sites of vitronectin expression, but failed to attach to normal brain. This adhesion was inhibited by antibodies directed against vitronectin, the alpha v beta 3 integrin, and with an Arg-Gly-Asp-containing peptide. These data provide evidence for a cell adhesion mechanism in glioblastoma tumors that might potentiate glioblastoma cell invasion of normal brain.


Asunto(s)
Glioblastoma/patología , Glicoproteínas/análisis , Receptores Inmunológicos/análisis , Animales , Adhesión Celular , Glioblastoma/química , Glicoproteínas/fisiología , Humanos , Invasividad Neoplásica , Conejos , Receptores de Vitronectina , Células Tumorales Cultivadas , Vitronectina
3.
J Clin Invest ; 96(3): 1621-30, 1995 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7544811

RESUMEN

Bleomycin-induced lung injury is an established murine model of human pulmonary fibrosis. Although procoagulant molecules (e.g., tissue factor [TF]) and fibrinolytic components (e.g., urokinase [u-PA] and type 1 plasminogen activator inhibitor [PAI-1]) have been detected in alveolar fluid from injured lungs, the origin of these molecules remains unknown. We therefore examined the expression of procoagulant and fibrinolytic components in relation to the distribution of parenchymal fibrin in bleomycin-injured lungs. Extravascular fibrin localized to the alveolar and extracellular matrix in injured lung tissue. Injured lung tissue extracts contained elevated levels of PAI-1 activity and decreased levels of u-PA activity. Whole lung PAI-1 and TF mRNAs were dramatically induced by lung injury. In situ hybridization of injured lungs revealed that PAI-1, u-PA, and TF mRNAs were induced within the fibrin-rich fibroproliferative lesions, primarily in fibroblast-like and macrophagelike cells, respectively, while TF mRNA was also induced in perilesional alveolar cells. Taken together, these observations suggest that the induction of PAI-1 and TF gene expression plays and important role in the formation and persistence of extracellular fibrin in bleomycin injured murine lungs.


Asunto(s)
Bleomicina/toxicidad , Expresión Génica , Pulmón/patología , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Tromboplastina/biosíntesis , Activador de Tejido Plasminógeno/biosíntesis , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Animales , Modelos Animales de Enfermedad , Femenino , Fibrina/análisis , Fibrina/biosíntesis , Fibrinólisis , Expresión Génica/efectos de los fármacos , Hibridación in Situ , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Factores de Tiempo
4.
Cancer Res ; 51(18): 4986-93, 1991 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-1893386

RESUMEN

Human glioblastomas (five of five), the most malignant astroglial-derived tumors, specifically express a chondroitin sulfate proteoglycan that is recognized by monoclonal antibody 9.2.27 and localized to the glioma cell surface, proliferating endothelial cells, and the perivascular extracellular matrix within the tumor bed. In contrast, the expression of this proteoglycan in normal adult neocortex and white matter is limited to the smooth muscle of small arteries, while normal glia, endothelial cells, and endothelial cell basement membranes are nonreactive. Moreover, two anaplastic astrocytomas, representing medium-grade astroglial-derived tumors, fail to react with monoclonal antibody 9.2.27. In culture, glioblastoma and capillary brain endothelial cells specifically synthesize a 250-kDa core protein and a high-molecular-mass chondroitin sulfate proteoglycan, recognized by monoclonal antibody 9.2.27. These data suggest a correlation between the expression of this chondroitin sulfate proteoglycan on proliferating brain capillary endothelial cells and the malignant phenotype of astroglial cells. The prominent perivascular localization of chondroitin sulfate proteoglycan makes it a marker for both proliferating brain capillary endothelial cells and the most malignant transformed astroglial cells, thus providing an ideal target for the immunotherapy of glioblastoma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/fisiología , Endotelio Vascular/metabolismo , Glioma/metabolismo , Meduloblastoma/metabolismo , Anticuerpos Monoclonales/inmunología , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Capilares/citología , Capilares/inmunología , Capilares/metabolismo , División Celular/fisiología , Proteoglicanos Tipo Condroitín Sulfato/biosíntesis , Proteoglicanos Tipo Condroitín Sulfato/inmunología , Endotelio Vascular/inmunología , Glioma/inmunología , Glioma/patología , Humanos , Meduloblastoma/inmunología , Meduloblastoma/patología , Fenotipo , Células Tumorales Cultivadas
5.
Cancer Res ; 59(9): 2076-82, 1999 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-10232591

RESUMEN

Anaplastic gliomas are infiltrative tumors, and their ability to migrate through normal brain contributes to their highly malignant behavior. Invasion of brain requires cell motility, which in turn depends on the activity of the cytoskeleton. A cytoskeletal component central to this process is myosin II, the cytoplasmic analogue of smooth and skeletal muscle myosin. Myosin II activity is regulated by the enzyme myosin light chain kinase, which activates myosin II by phosphorylating it on its regulatory light chain. We have investigated the role of myosin II in glioma motility and invasiveness by examining the effects of two inhibitors of myosin light chain kinase, ML7 and KT5926. Both drugs are potent inhibitors of both glioma motility, as measured by a scrape motility assay, and an in vitro haptotaxis assay. The inhibition of in vitro haptotaxis follows the dose-response relationship expected for competitive inhibition of myosin light chain kinase by these drugs and is seen at drug concentrations that are nontoxic. These results highlight the important role that myosin II contributes to glioma invasiveness and suggest that it may serve as a target in future strategies at blocking invasion by these tumors.


Asunto(s)
Alcaloides/farmacología , Azepinas/farmacología , Carbazoles , Glioma/patología , Indoles , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Miosinas/fisiología , Naftalenos/farmacología , Invasividad Neoplásica , Proteínas de Neoplasias/antagonistas & inhibidores , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/ultraestructura , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Integrinas/análisis , Microscopía Fluorescente , Fosforilación/efectos de los fármacos , Células Tumorales Cultivadas/efectos de los fármacos
6.
Cancer Res ; 59(14): 3369-73, 1999 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10416596

RESUMEN

The urokinase-type plasminogen activator (uPA) and uPA receptor (UPAR) play important roles in the proteolytic cascade involved in the invasiveness of gliomas and other invasive tumors. High-level expression of uPAR has been correlated with high-grade glioma cell lines and tumors We report here that down-regulating uPAR levels by antisense strategy using an adenovirus construct (Ad-uPAR) inhibited glioma invasion in Matrigel and spheroid in vitro models. sc. (U87-MG) and intracranial (SNB19) injections of Ad-uPAR-infected glioma cells did not produce tumors in nude mice. However, injection of the Ad-uPAR construct into previously established so U87-MG tumors in nude mice caused regression of those tumors. Our results support the therapeutic potential of targeting the uPA-uPAR system for the treatment of gliomas and other cancers.


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/terapia , ADN sin Sentido/genética , Terapia Genética , Glioblastoma/terapia , Proteínas de Neoplasias/genética , Receptores de Superficie Celular/genética , Animales , Neoplasias Encefálicas/patología , Progresión de la Enfermedad , Glioblastoma/patología , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica , Proteínas de Neoplasias/antagonistas & inhibidores , Trasplante de Neoplasias , Organoides , Ratas , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Células Tumorales Cultivadas
7.
Oncogene ; 19(50): 5801-9, 2000 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-11126367

RESUMEN

Synthesis of cell attachment proteins and cytokines, such as osteopontin (OPN), can promote tumor cell remodeling of the extracellular matrix into an environment that promotes tumor cell attachment and migration. We investigated the transcriptional regulation of OPN in the U-251MG and U-87MG human malignant astrocytoma cell lines. Deletion and mutagenesis analyses of the OPN promoter region identified a proximal promoter element (-24 to -94 relative to the transcription initiation site) that is essential for maintaining high levels of OPN expression in the tumor cells. This element, designated RE-1, consists of two cis-acting elements, RE-1a (-55 to -86) and RE-1b (-22 to -45), which act synergistically to regulate the activity of the OPN promoter. Gel shift assays using nuclear extracts of U-251MG cells demonstrated that RE-1a contains binding sites for transcription factors Sp1, the glucocorticoid receptor, and the E-box-binding factors, whereas RE-1b contains a binding site for the octamer motif-binding protein (OCT-1/OCT-2). Inclusion of antibodies directed toward Myc and OCT-1 in the gel shift assays indicated that Myc and OCT-1 participate in forming DNA-protein complexes on the RE-1a and RE-1b elements, respectively. Our results identify two previously unrecognized elements in the OPN promoter that act synergistically to promote upregulation of OPN synthesis by tumor cells but are regulated by different transcription factors.


Asunto(s)
ADN de Neoplasias/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Regiones Promotoras Genéticas/genética , Sialoglicoproteínas/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Anticuerpos Monoclonales/farmacología , Astrocitos/metabolismo , Astrocitos/fisiología , Astrocitoma/genética , Astrocitoma/metabolismo , Sitios de Unión , ADN de Neoplasias/genética , Eliminación de Gen , Factor C1 de la Célula Huésped , Humanos , Mutagénesis Sitio-Dirigida , Factor 1 de Transcripción de Unión a Octámeros , Factor 2 de Transcripción de Unión a Octámeros , Osteopontina , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/inmunología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Secuencias Reguladoras de Ácidos Nucleicos , Sialoglicoproteínas/biosíntesis , Factor de Transcripción Sp1/metabolismo , Células Tumorales Cultivadas
8.
Oncogene ; 20(55): 7976-86, 2001 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-11753680

RESUMEN

Cytokines can influence the interactions between members of the integrin cell adhesion receptor family and the extracellular matrix thereby potentially affecting cell function and promoting cell adhesion, growth and migration of malignant astrocytoma tumor cells. As malignant astrocytoma cells synthesize TGF-beta1 in vivo, we analysed the effects of TGF-beta1 on signaling events associated with integrin receptor ligation, focusing on the effects on paxillin, a phosphorylated adaptor protein, that acts as a scaffold for signaling molecules recruited to focal adhesions. TGF-beta1-stimulation of primary astrocytes and serum-starved U-251MG malignant astrocytoma cells attached to fibronectin induced a substantial increase in the levels of paxillin protein (fivefold increase at 2.0 ng/ml) in a dose- and time-dependent manner compared to the levels observed on plating onto fibronectin in the absence of stimulation. In the astrocytoma cells, this resulted in an increase in the pool of tyrosine-phosphorylated paxillin, although it did not appear to alter the extent of phosphorylation of the paxillin molecules. In contrast, in primary astrocytes the protein levels were upregulated in the absence of a parallel increase in phosphorylation. The TGF-beta1-stimulated increase in paxillin levels required ligation of the fibronectin receptor, as it was not induced when the cells were plated onto vitronectin, collagen or laminin. The increase in the pool of paxillin on TGF-beta1 stimulation of the fibronectin-plated astrocytoma cells was associated with an increase in translation, but was not associated with an increase in the steady-state levels of paxillin mRNA. Stimulation with TGF-beta1 on a fibronectin substrate increased subsequent attachment and spreading of U-251MG cells onto fibronectin and, to a lesser extent, vitronectin, but not collagen. Our results indicate that physiologic levels of TGF-beta1 stimulate the expression of paxillin protein at the level of translation through a process that requires engagement of the fibronectin receptor, and promotes attachment and spreading of malignant astrocytoma cells on fibronectin.


Asunto(s)
Astrocitoma/metabolismo , Proteínas del Citoesqueleto/metabolismo , Fibronectinas/metabolismo , Fosfoproteínas/metabolismo , Receptores de Fibronectina/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba/efectos de los fármacos , Animales , Western Blotting , Adhesión Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Neoplasias del Sistema Nervioso Central/metabolismo , Proteínas del Citoesqueleto/biosíntesis , Relación Dosis-Respuesta a Droga , Matriz Extracelular/metabolismo , Semivida , Humanos , Microscopía Fluorescente , Paxillin , Fosfoproteínas/biosíntesis , Fosforilación/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Seudópodos/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Especificidad por Sustrato , Factores de Tiempo , Factor de Crecimiento Transformador beta1 , Células Tumorales Cultivadas
9.
Clin Cancer Res ; 5(6): 1587-94, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10389948

RESUMEN

Vitronectin (VN) is an extracellular matrix (ECM) protein, the synthesis of which in vivo by glioma cells correlates with tumor grade. Although the role of VN as a permissive substrate for glioma migration has been well characterized, its role in conferring a survival advantage for tumor cells has not been addressed previously. By using an in vitro assay of DNA fragmentation as a quantitative measure of apoptotic cell death, we sought to determine whether the sensitivity of two human glioma cell lines (D54 and U251) to drug-induced apoptosis could be inhibited by VN. As well, the extent to which apoptosis could be inhibited was correlated with the levels of the Bcl-2 family of proteins that are known to modulate apoptosis and chemoresistance. Results of the study were: (a) VN coatings, in a dose-dependent manner, inhibited topoisomerase (Topo)-induced apoptosis by up to 50% (optimal coating density, 500 ng/cm2); in contrast, fibronectin (FN), an ECM protein present in abundance in the brain, demonstrated no protection; (b) in a dose-response study, VN clearly conferred a survival advantage (LD50 of Topo: on VN, 120 ng/ml; on FN, 35 ng/ml); (c) the protective effect of VN was not due to enhanced cell adhesion or alterations in the cell cycle distribution; (d) both of the classic integrin receptors that bind VN (alpha(v)beta3, alpha(v)beta5) were capable of mediating this protective effect, because ligation of either of the two classic integrins conferred chemoresistance to Topo; and (e) chemoresistance observed with VN was associated with an increase in expression of two antiapoptotic proteins, Bcl-2 and Bcl-X(L), with a consequent increase in the ratios for Bcl-2:Bax and Bcl-X(L):Bax. VN, an ECM protein preferentially expressed at the tumor-brain interface in vivo, may confer a survival advantage to glioma cells at the advancing tumor margin and may thus, in part, underlie the high level of tumor recurrence at this interface.


Asunto(s)
Apoptosis , Glioma/patología , Vitronectina/fisiología , Anticuerpos Monoclonales/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Fibronectinas/farmacología , Humanos , Etiquetado Corte-Fin in Situ , Integrinas/antagonistas & inhibidores , Integrinas/inmunología , Integrinas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Receptores de Fibronectina/antagonistas & inhibidores , Receptores de Fibronectina/inmunología , Receptores de Fibronectina/fisiología , Receptores de Vitronectina/antagonistas & inhibidores , Receptores de Vitronectina/inmunología , Receptores de Vitronectina/fisiología , Topotecan/farmacología , Células Tumorales Cultivadas , Proteína bcl-X
10.
J Neuropathol Exp Neurol ; 55(11): 1143-9, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8939197

RESUMEN

Angiogenesis, which promotes tumor proliferation and invasiveness, is mediated by integrin-dependent cell adhesion mechanisms and requires expression of integrin alpha v beta 3. To determine whether integrin alpha v beta 3 is expressed in the small blood vessel hyperplasia characteristic of glioblastoma tumors, we explored the mRNA and protein expression of integrin alpha v, beta 3, beta 5, and beta 1 subunits in small blood vessels in gliomas of various grades and nontumorous brain biopsies. Antisense beta 3 and beta 5 riboprobes hybridized to small blood vessels (endothelial and adjacent mesenchymal cells) in 11 of 11 glioblastomas and 2 of 4 anaplastic astrocytomas, but failed to hybridize to small blood vessels in 12 non-neoplastic specimens and 4 low-grade astrocytomas. In contrast, antisense alpha v and beta 1 riboprobes hybridized to small blood vessels in all of the biopsies. The expression of integrin alpha v beta 3 protein in these vessels was determined by immunohistochemical analysis using anti-integrin subunit-specific antibodies. The findings were similar to those obtained using in situ hybridization: beta 3 subunit protein was detected on small blood vessels in 9 of 12 glioblastoma and 2 of 4 anaplastic astrocytomas, whereas it was not detected in small blood vessels of 10 non-neoplastic brains and 4 low-grade astrocytomas. In contrast, the alpha v and beta 1 subunit proteins were detected in small blood vessels in all of the brain biopsies, whereas the beta 5 subunit protein could not be detected. These data suggest that integrin alpha v beta 3 is expressed in small blood vessels of glioblastoma tumors.


Asunto(s)
Neoplasias Encefálicas/irrigación sanguínea , Glioblastoma/irrigación sanguínea , Receptores de Vitronectina/metabolismo , Adulto , Anciano , Vasos Sanguíneos/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Humanos , Inmunohistoquímica , Hibridación in Situ , Persona de Mediana Edad , ARN Mensajero/metabolismo , Receptores de Vitronectina/genética
11.
J Neuropathol Exp Neurol ; 58(10): 1029-40, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10515226

RESUMEN

The ECM of astrocytic tumors promotes and modulates a variety of cell functions, such as cell attachment, migration, proliferation, survival, and signaling. Recent studies indicate that there are extensive and complex interactions among ECM molecules and that these can modify the function of the participating molecules, interactions between the proteoglycan, phosphacan, and the ECM protein, tenascin, being an example (63). In addition, on nonastrocytic cell types it has been shown that an integrin receptor and the cell surface proteoglycan CD44 recognize the same ECM ligand osteopontin, and thus modulate each others function (77, 86). Thus, interacting components in the ECM and cell surface receptors likely cooperate in regulating cell function and tumor invasion (59, 77, 80, 85-87, 95). As tumor cells are capable of remodeling their ECM through synthesis of ECM proteins and proteoglycans, as well as upregulating integrin receptors and proteoglycans on their cell surface, tumor cells are capable of controlling their own destiny. ECM molecules which are concentrated in blood vessels of malignant astrocytomas, such as tenascin-C and the 250-kDa CSPG (NG2), are potentially therapeutic targets.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Matriz Extracelular/metabolismo , Glioma/metabolismo , Vasos Sanguíneos/metabolismo , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/fisiopatología , Proteínas de la Matriz Extracelular/metabolismo , Glioma/irrigación sanguínea , Glioma/patología , Glioma/fisiopatología , Humanos , Receptores de Superficie Celular/metabolismo
12.
J Neuropathol Exp Neurol ; 58(9): 972-81, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10499439

RESUMEN

Hypoxia associated with perinatal events can result in brain damage in the neonate. In labor and eclampsia, hypoxia can be intermittent, which may result in more severe damage than sustained hypoxia. The pathogenesis of brain injury in sustained ischemia involves free radical production; therefore, we investigated whether higher levels of free radicals contribute to the greater injury induced by repetitive ischemia. Brains were obtained from fetuses of near-term, pregnant rabbits subjected to repetitive ischemia-reperfusion (RIR), sustained uterine ischemia-reperfusion (IR), or a control protocol. Compared with controls, fetal brains from RIR or IR groups had more brain edema. Brains from RIR fetuses exhibited higher levels of lipid peroxidation, 3-nitrotyrosine, and nitrogen oxides, and lower total antioxidant capacity and cortical cellular viability than those of IR or control fetuses. Maternal administration of antioxidants following RIR and fetal bradycardia resulted in lower levels of fetal cortical and hippocampal cell death. Coadministration of Trolox and ascorbic acid resulted in less brain edema and liquefaction, and fewer hippocampal ischemic nuclei as compared with the saline control. Higher free radical production may be responsible for the greater fetal brain injury following repetitive hypoxia-reoxygenation. Maternal antioxidant treatment resulted in transplacental passage of antioxidants and amelioration of brain injury, and may be a viable clinical option following diagnosis of fetal distress.


Asunto(s)
Encéfalo/embriología , Enfermedades Fetales/metabolismo , Enfermedades Fetales/patología , Radicales Libres/metabolismo , Hipoxia/metabolismo , Hipoxia/patología , Oxígeno/farmacología , Animales , Antioxidantes/farmacología , Isquemia Encefálica/patología , Femenino , Feto/metabolismo , Nitrógeno/metabolismo , Estrés Oxidativo , Embarazo , Conejos , Daño por Reperfusión/patología
13.
J Neuropathol Exp Neurol ; 57(6): 544-53, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9630234

RESUMEN

Free radical-mediated injury is implicated in hypoxic-ischemic encephalopathy observed in neonates. We investigated in utero free radical production and injury following hypoxia-ischemia to premature fetal brain utilizing a rabbit model of acute placental insufficiency. Pregnant rabbits at 29 days gestation were randomized to uterine ischemia for 50 minutes (min) (hypoxia) or nonischemic controls. Fetal brains were obtained immediately after ischemia for oxidative and acute-injury markers or 24 hours (h) post-ischemia for histopathology. Nitrotyrosine formation, a marker of NO-derived species such as peroxynitrite, was observed only in hypoxic brains. Hypoxia resulted in a significant increase in nitrogen oxides, lipid peroxidation, and protein oxidation, with a concomitant decrease in total antioxidant capacity, compared with controls. Peroxynitrite addition to brain homogenate increased nitrogen oxides linearly (1:1), although protein carbonyls were unchanged. Concomitantly, in vitro cortical and hippocampal cell viability and ATP levels decreased, with an increase in brain edema in hypoxic brains. Fetuses delivered 24 h post-ischemia had increased hippocampal nuclear karyorrhexis on histology compared with controls. Antioxidant administration (ascorbic acid and Trolox) intraperitoneally ameliorated changes in cellular viability and brain edema. Acute fetal hypoxia-ischemia without reoxygenation results in increased nitrogen and oxygen free radical production that may cause brain injury. The merits of the described model are discussed.


Asunto(s)
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Hipoxia Encefálica/metabolismo , Nitrógeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Encéfalo/citología , Encéfalo/embriología , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromanos/farmacología , Femenino , Radicales Libres/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Embarazo , Conejos , Útero/irrigación sanguínea
14.
Neurology ; 41(1): 124-8, 1991 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1985277

RESUMEN

We describe a term newborn with tuberous sclerosis who presented with a neonatal brain tumor, diagnosed as a subependymal giant cell astrocytoma. We compare the various imaging modalities used in the diagnosis of this tumor.


Asunto(s)
Neoplasias del Ventrículo Cerebral/complicaciones , Glioma/complicaciones , Esclerosis Tuberosa/complicaciones , Neoplasias del Ventrículo Cerebral/diagnóstico , Ventrículos Cerebrales/patología , Ventriculografía Cerebral , Glioma/diagnóstico , Humanos , Recién Nacido , Imagen por Resonancia Magnética , Masculino , Tomografía Computarizada por Rayos X , Ultrasonografía
15.
Front Biosci ; 4: D178-87, 1999 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9989951

RESUMEN

The urokinase-type plasminogen activator receptor (uPAR) plays a critical role in the regulation of cell-surface plasminogen activation in several physiological and pathological conditions. Recent evidence suggests that the uPAR is also involved in processes that are not related to plasminogen activation, including cell adhesion and transmission of extracellular signals across the plasma membrane. The uPAR influences cell migration and spreading both in vivo and in vitro through the cell-surface activation of plasminogen. The uPAR can bind to vitronectin, an adhesive extracellular matrix protein that contains the Arg-gly-Asp (RGD) cell adhesion domain and that serves as a ligand for several integrin receptors. uPAR also forms complexes with (1, (2, and (3 integrins, thereby allowing mutual interactions and regulation between cell adhesion and proteolysis. Recently, uPAR has been shown to have strong prognostic value for predicting disease recurrence and overall survival in certain types of cancer. We discuss here the biological significance of uPAR in the glioblastoma invasion process. Strong correlations found between elevated uPAR levels in glioblastoma cells and tumor invasiveness have led to uPAR being selected as a target for therapy in experimental animal models. Using antisense vectors to down regulate uPAR expression at the level of the mRNA and protein in glioblastoma cells, has been shown to inhibit tumor formation in nude mice. These results provide a potential basis from which to develop novel therapeutic strategies to direct the expression of antisense uPAR and to evaluate the efficiency of this technique for cancer gene therapy in patients with brain tumor.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Activadores Plasminogénicos/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Adhesión Celular , Humanos , Invasividad Neoplásica , Activadores Plasminogénicos/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
16.
Front Biosci ; 4: D188-99, 1999 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9989953

RESUMEN

Integrins are cell surface receptors that mediate the physical and functional interactions between a cell and its surrounding extracellular matrix (ECM). Expressed as heterodimers, the specific alpha or beta chains that constitute the integrin receptor determine the repertoire of ECM proteins to which a specific integrin may bind (table 1). While classically, the role ascribed to integrins has been that of anchoring cells to the ECM, the more contemporary spectrum of integrin function greatly exceeds that of mere cell adhesion. Recent reports have demonstrated that the interaction between the ECM and cell surface integrins leads to intracellular signaling events that affect cell migration, proliferation, and survival, which in the context of neoplastic cells, can translate directly into the malignant phenotype (1). Indeed, the role of specific integrins in tumorigenesis has been demonstrated in numerous cancer types (table 2). In primary tumors of the nervous system, the contribution of integrins to the malignant phenotype of gliomas has been an area of significant attention and research in numerous laboratories, including that of ours. As illustrated in table 3, several integrins have been identified as being of key importance in glioma biology. In this article, we review the current knowledge of how these integrins influence the malignant characteristics of gliomas and, as such, how these cell surface receptors may thus represent potential targets in the design of future therapeutics for patients afflicted with gliomas.


Asunto(s)
Glioma/patología , Integrinas/fisiología , Animales , División Celular , Movimiento Celular , Supervivencia Celular , Resistencia a Antineoplásicos , Endopeptidasas/metabolismo , Proteínas de la Matriz Extracelular/fisiología , Fibronectinas/metabolismo , Humanos , Integrina beta1/fisiología , Laminina/metabolismo , Invasividad Neoplásica , Neovascularización Patológica , Fenotipo , Tenascina/fisiología , Vitronectina/fisiología
17.
Am J Surg Pathol ; 15(3): 222-6, 1991 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1996728

RESUMEN

Skeletal muscle biopsies from five patients with severe myalgias, peripheral eosinophilia, and a recent history of L-tryptophan ingestion were analyzed. Perimysial inflammation, predominantly mononuclear with variable numbers of eosinophils was seen (in five of five patients), which was perineurial (in three of five) and perivascular (in five of five) in location. Grouping of the myofiber types was identified by enzyme histochemistry in two of four patients; fresh muscle for histochemical studies was unavailable from one patient. An occasional degenerating myofiber was seen in only one patient, who was still ingesting L-tryptophan at the time of biopsy. No vasculitis was seen. The focus of muscle injury in this syndrome appeared to be the perimysium and, in particular, the perineurial and perivascular connective tissue.


Asunto(s)
Eosinofilia/inducido químicamente , Músculos/enzimología , Enfermedades Musculares/inducido químicamente , Miositis/inducido químicamente , Neuritis/inducido químicamente , Triptófano/efectos adversos , Adulto , Anciano , Biopsia , Eosinofilia/enzimología , Eosinofilia/patología , Femenino , Histocitoquímica , Humanos , Masculino , Persona de Mediana Edad , Músculos/patología , Enfermedades Musculares/enzimología , Enfermedades Musculares/patología , Síndrome
18.
Thromb Haemost ; 59(1): 18-22, 1988 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-2966450

RESUMEN

The frequency of heterozygous protein C and protein S deficiency, detected by measuring total plasma antigen, in a group (n = 141) of young unrelated patients (less than 45 years old) with venous thrombotic disease was studied and compared to that of antithrombin III, fibrinogen, and plasminogen deficiencies. Among 91 patients not receiving oral anticoagulants, six had low protein S antigen levels and one had a low protein C antigen level. Among 50 patients receiving oral anticoagulant therapy, abnormally low ratios of protein S or C to other vitamin K-dependent factors were presented by one patient for protein S and five for protein C. Thus, heterozygous Type I protein S deficiency appeared in seven of 141 patients (5%) and heterozygous Type I protein C deficiency in six of 141 patients (4%). Eleven of thirteen deficient patients had recurrent venous thrombosis. In this group of 141 patients, 1% had an identifiable fibrinogen abnormality, 2% a plasminogen abnormality, and 3% an antithrombin III deficiency. Thus, among the known plasma protein deficiencies associated with venous thrombosis, protein S and protein C deficiencies (9%) emerge as the leading identifiable associated abnormalities.


Asunto(s)
Glicoproteínas/deficiencia , Heterocigoto , Deficiencia de Proteína C , Tromboflebitis/genética , Adulto , Anticoagulantes/uso terapéutico , Femenino , Glicoproteínas/genética , Humanos , Masculino , Proteína C/genética , Proteína S , Tromboflebitis/sangre
19.
Arch Dermatol ; 123(12): 1701a-1706a, 1987 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2961308

RESUMEN

Protein C (PC), a 62,000-molecular weight vitamin K-dependent serine protease zymogen, is a natural anticoagulant that occurs in plasma at 4 mg/L. Activated PC inactivates clotting factors V and VIII and is also profibrinolytic. Activated PC is enhanced in its anticoagulant activity by protein S (PS), another vitamin K-dependent protein. Protein S is found in platelets and endothelial cells as well as in plasma. Inherited PC deficiency and PS deficiency have been associated with venous thrombosis. Both heterozygous PC and PS deficiency appear to be inherited in an autosomal dominant manner in some families. Homozygous PC deficiency presents as neonatal purpura fulminans and results in massive venous thrombosis of the skin and other organs within the first few days of life. Symptomatic heterozygous PC deficiency and PS deficiency have been treated with oral anticoagulants, successfully minimizing recurrence of thrombosis. Coumarin-induced skin necrosis, a rare complication of oral anticoagulant therapy usually seen within three to five days of initiation of therapy, has also been associated with heterozygous PC deficiency. The short half-life of PC (six to eight hours) compared with most of the vitamin K-dependent clotting factors (greater than 30 hours) is the probable reason for this paradoxical response to oral anticoagulants in some PC-deficient patients, since a transient imbalance of procoagulant and anticoagulant factors may exist during initiation of oral anticoagulant therapy. Acquired deficiency of the PC pathway occurs in disseminated intravascular coagulation and possibly other diseases such as those associated with a lupus anticoagulant.


Asunto(s)
Cumarinas/efectos adversos , Deficiencia de Proteína C , Púrpura/etiología , Piel/efectos de los fármacos , Adolescente , Adulto , Glicoproteínas/deficiencia , Glicoproteínas/fisiología , Heterocigoto , Homocigoto , Humanos , Recién Nacido , Necrosis , Proteína C/análisis , Proteína C/fisiología , Proteína S , Púrpura/tratamiento farmacológico , Púrpura/genética , Piel/patología
20.
Neurosci Lett ; 283(2): 157-61, 2000 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-10739899

RESUMEN

Vitronectin is expressed in a cell-specific manner in the developing brain and concentrated in the brain during disease processes, such as germinal matrix hemorrhage and infarction, in which there is breakdown of the blood-brain barrier. In this study, we identified the integrin receptors that mediate attachment of primary neonatal rat astrocytes to vitronectin. Using fluorescent activated cell sorter and immunoprecipitation analyses, we established that the vitronectin receptor integrins alphavbeta5 and alpha8beta1, but not alphavbeta3, are expressed on neonatal rat astrocytes. Attachment of the neonatal astrocytes to vitronectin was inhibited (85%) in an additive manner by neutralizing anti-alphavbeta5 and anti-beta1 antibodies. Attachment to vitronectin was also inhibited in a dose-dependent manner by the type I plasminogen activator inhibitor (PAI-1), a serine protease inhibitor. Our data demonstrate that unstimulated primary neonatal rat astrocytes attach to vitronectin, utilizing integrins alphavbeta5 and alpha8beta1, and that this attachment is regulated by PAI-1.


Asunto(s)
Astrocitos/fisiología , Adhesión Celular , Integrinas/fisiología , Inhibidor 1 de Activador Plasminogénico/farmacología , Vitronectina , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/efectos de los fármacos , Encéfalo/citología , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Ratas , Receptores de Fibronectina/fisiología , Receptores de Vitronectina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA