Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Pathol ; 232(3): 369-81, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24421076

RESUMEN

Although the incidence of breast cancer metastasis (BCM) in brain has increased significantly in triple-negative breast cancer (TNBC), the mechanisms remain elusive. Using in vivo mouse models for BCM in brain, we observed that TNBC cells crossed the blood-brain barrier (BBB), lodged in the brain microvasculature and remained adjacent to brain microvascular endothelial cells (BMECs). Breaching of the BBB in vivo by TNBCs resulted in increased BBB permeability and changes in ZO-1 and claudin-5 tight junction (TJ) protein structures. Angiopoietin-2 expression was elevated in BMECs and was correlated with BBB disruption. Secreted Ang-2 impaired TJ structures and increased BBB permeability. Treatment of mice with the neutralizing Ang-2 peptibody trebananib prevented changes in the BBB integrity and BMEC destabilization, resulting in inhibition of TNBC colonization in brain. Thus, Ang-2 is involved in initial steps of brain metastasis cascade, and inhibitors for Ang-2 may serve as potential therapeutics for brain metastasis.


Asunto(s)
Angiopoyetina 2/metabolismo , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Barrera Hematoencefálica/patología , Permeabilidad Capilar/fisiología , Femenino , Xenoinjertos , Humanos , Inmunohistoquímica , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Neoplasias de la Mama Triple Negativas/patología
2.
Int J Cancer ; 134(5): 1034-44, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23934616

RESUMEN

Neuropeptide substance P (SP) has been implicated in inflammation, pain, depression and breast cancer cell (BCC) growth. Here, we examined the role of SP in trafficking of BCCs (human MDA-MB-231 and MDA-MB-231BrM2 cells) across the blood-brain barrier (BBB) and brain microvascular endothelial cells (BMECs) using in vitro and in vivo models. SP was secreted from BCCs and mediated adhesion and transmigration of BCCs across human BMECs (HBMECs) in vitro. SP induced activation of HBMECs, leading to secretion of Tumor Necrosis Factor alpha (TNF-α) and angiopoietin-2 (Ang-2) from HBMECs, resulting in changes in localization and distribution of tight junction (TJ) ZO-1 (tight junction protein zonula occludins-1) and claudin-5 structures as well as increased permeability of HBMECs. Using spontaneous breast cancer metastasis mouse model (syngeneic) of GFP-4T1-BrM5 mammary tumor cells administered into mammary fat pads of Balb/c mice, SP inhibitor spantide III inhibited in vivo changes in permeability of the BBB and BMEC-TJs ZO-1 and claudin-5 structures as well as decreased tumor cell colonization in brain. Thus, SP secreted from BCCs induces transmigration of BCCs across the BBB, leading to activation of BMECs and secretion of TNF-α and Ang-2, resulting in BBB impairment and colonization of tumor cells in brain. Therefore, therapies based on SP inhibition in combination with other therapies may prevent breaching of the BBB by BCCs and their colonization in brain.


Asunto(s)
Barrera Hematoencefálica , Neoplasias de la Mama/patología , Células Endoteliales/fisiología , Sustancia P/fisiología , Uniones Estrechas/fisiología , Animales , Adhesión Celular , Movimiento Celular , Citoesqueleto/química , Femenino , Humanos , Ratones , Microvasos/citología , Permeabilidad , Sustancia P/análogos & derivados , Sustancia P/antagonistas & inhibidores , Sustancia P/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
3.
Blood ; 117(3): 827-38, 2011 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21063029

RESUMEN

Endocannabinoids are arachidonic acid derivatives and part of a novel bioactive lipid signaling system, along with their G-coupled cannabinoid receptors (CB1 and CB2) and the enzymes involved in their biosynthesis and degradation. However, their roles in hematopoiesis and hematopoietic stem and progenitor cell (HSPC) functions are not well characterized. Here, we show that bone marrow stromal cells express endocannabinoids (anandamide and 2-arachidonylglycerol), whereas CB2 receptors are expressed in human and murine HSPCs. On ligand stimulation with CB2 agonists, CB2 receptors induced chemotaxis, migration, and enhanced colony formation of bone marrow cells, which were mediated via ERK, PI3-kinase, and Gαi-Rac1 pathways. In vivo, the CB2 agonist AM1241 induced mobilization of murine HSPCs with short- and long-term repopulating abilities. In addition, granulocyte colony-stimulating factor -induced mobilization of HSPCs was significantly decreased by specific CB2 antagonists and was impaired in Cnr2(-/-) cannabinoid type 2 receptor knockout mice. Taken together, these results demonstrate that the endocannabinoid system is involved in hematopoiesis and that CB2/CB2 agonist axis mediates repopulation of hematopoiesis and mobilization of HSPCs. Thus, CB2 agonists may be therapeutically applied in clinical conditions, such as bone marrow transplantation.


Asunto(s)
Hematopoyesis/fisiología , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/fisiología , Receptor Cannabinoide CB2/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Moduladores de Receptores de Cannabinoides/metabolismo , Cannabinoides/farmacología , Movimiento Celular/efectos de los fármacos , Ciclohexanoles/farmacología , Femenino , Citometría de Flujo , Hematopoyesis/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/genética , Células del Estroma/metabolismo
4.
J Biol Chem ; 285(34): 26190-8, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20511222

RESUMEN

Reactive molecules have diverse effects on cells and contribute to several pathological conditions. Cells have evolved complex protective systems to neutralize these molecules and restore redox homeostasis. Previously, we showed that association of nuclear factor (NF)-erythroid-derived 2 (E2)-related factor 2 (NRF2) with the nuclear matrix protein NRP/B was essential for the transcriptional activity of NRF2 target genes in tumor cells. The present study demonstrates the molecular mechanism by which NRP/B, via NRF2, modulates the transcriptional activity of antioxidant response element (ARE)-driven genes. NRP/B is localized in the nucleus of primary brain tissue and human neuroblastoma (SH-SY5Y) cells. Treatment with hydrogen peroxide (H(2)O(2)) enhances the nuclear colocalization of NRF2 and NRP/B and induces heme oxygenase 1 (HO1). Treatment of NRP/B or NRF2 knockdowns with H(2)O(2) induced apoptosis. Co-expression of NRF2 with members of the Kelch protein family, NRP/B, MAYVEN, or MAYVEN-related protein 2 (MRP2), revealed that the NRF2-NRP/B complex is important for the transcriptional activity of ARE-driven genes HO1 and NAD(P)H:quinine oxidoreductase 1 (NQO1). NRP/B interaction with Nrf2 was mapped to NRF2 ECH homology 4 (Neh4)/Neh5 regions of NRF2. NRP/B mutations that resulted in low binding affinity to NRF2 were unable to activate NRF2-modulated transcriptional activity of the ARE-driven genes, HO1 and NQO1. Thus, the interaction of NRP/B with the Neh4/Neh5 domains of NRF2 is indispensable for activation of NRF2-mediated ARE-driven antioxidant and detoxifying genes that confer cellular defense against oxidative stress-induced damage.


Asunto(s)
Proteínas de Microfilamentos/fisiología , Factor 2 Relacionado con NF-E2/metabolismo , Neuropéptidos/fisiología , Proteínas Asociadas a Matriz Nuclear/fisiología , Proteínas Nucleares/fisiología , Estrés Oxidativo/genética , Activación Transcripcional , Antioxidantes , Química Encefálica , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Neuroblastoma/química , Oxidación-Reducción , Elementos de Respuesta
5.
J Biol Chem ; 285(46): 35471-8, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20826813

RESUMEN

Endocannabinoids are lipid signaling molecules that act via G-coupled receptors, CB(1) and CB(2). The endocannabinoid system is capable of activation of distinct signaling pathways on demand in response to pathogenic events or stimuli, hereby enhancing cell survival and promoting tissue repair. However, the role of endocannabinoids in hematopoietic stem and progenitor cells (HSPCs) and their interaction with hematopoietic stem cells (HSC) niches is not known. HSPCs are maintained in the quiescent state in bone marrow (BM) niches by intrinsic and extrinsic signaling. We report that HSPCs express the CB(1) receptors and that BM stromal cells secrete endocannabinoids, anandamide (AEA) (35 pg/10(7) cells), and 2-AG (75.2 ng/10(7) cells). In response to the endotoxin lipopolysaccharide (LPS), elevated levels of AEA (75.6 pg/10(7) cells) and 2-AG (98.8 ng/10(7) cells) were secreted from BM stromal cells, resulting in migration and trafficking of HSPCs from the BM niches to the peripheral blood. Furthermore, administration of exogenous cannabinoid CB(1) agonists in vivo induced chemotaxis, migration, and mobilization of human and murine HSPCs. Cannabinoid receptor knock-out mice Cnr1(-/-) showed a decrease in side population (SP) cells, whereas fatty acid amide hydrolase (FAAH)(-/-) mice, which have elevated levels of AEA, yielded increased colony formation as compared with WT mice. In addition, G-CSF-induced mobilization in vivo was modulated by endocannabinoids and was inhibited by specific cannabinoid antagonists as well as impaired in cannabinoid receptor knock-out mice Cnr1(-/-), as compared with WT mice. Thus, we propose a novel function of the endocannabinoid system, as a regulator of HSPC interactions with their BM niches, where endocannabinoids are expressed in HSC niches and under stress conditions, endocannabinoid expression levels are enhanced to induce HSPC migration for proper hematopoiesis.


Asunto(s)
Células de la Médula Ósea/metabolismo , Moduladores de Receptores de Cannabinoides/biosíntesis , Endocannabinoides , Células Madre Hematopoyéticas/metabolismo , Células del Estroma/metabolismo , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Ácidos Araquidónicos/biosíntesis , Western Blotting , Células de la Médula Ósea/citología , Moduladores de Receptores de Cannabinoides/fisiología , Comunicación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Ciclohexanoles/farmacología , Femenino , Citometría de Flujo , Glicéridos/biosíntesis , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Alcamidas Poliinsaturadas , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/genética , Receptor Cannabinoide CB2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Población Lateral/citología , Células de Población Lateral/metabolismo , Nicho de Células Madre/citología , Nicho de Células Madre/metabolismo , Células del Estroma/citología
6.
J Immunol ; 181(9): 6406-16, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18941231

RESUMEN

HIV-1 infection has significant effect on the immune system as well as on the nervous system. Breakdown of the blood-brain barrier (BBB) is frequently observed in patients with HIV-associated dementia (HAD) despite lack of productive infection of human brain microvascular endothelial cells (HBMEC). Cellular products and viral proteins secreted by HIV-1 infected cells, such as the HIV-1 Gp120 envelope glycoprotein, play important roles in BBB impairment and HIV-associated dementia development. HBMEC are a major component of the BBB. Using cocultures of HBMEC and human astrocytes as a model system for human BBB as well as in vivo model, we show for the first time that cannabinoid agonists inhibited HIV-1 Gp120-induced calcium influx mediated by substance P and significantly decreased the permeability of HBMEC as well as prevented tight junction protein down-regulation of ZO-1, claudin-5, and JAM-1 in HBMEC. Furthermore, cannabinoid agonists inhibited the transmigration of human monocytes across the BBB and blocked the BBB permeability in vivo. These results demonstrate that cannabinoid agonists are able to restore the integrity of HBMEC and the BBB following insults by HIV-1 Gp120. These studies may lead to better strategies for treatment modalities targeted to the BBB following HIV-1 infection of the brain based on cannabinoid pharmacotherapies.


Asunto(s)
Fármacos Anti-VIH/farmacología , Ácidos Araquidónicos/farmacología , Encéfalo/irrigación sanguínea , Encéfalo/virología , Moduladores de Receptores de Cannabinoides/farmacología , Endotelio Vascular/virología , Glicéridos/farmacología , Proteína gp120 de Envoltorio del VIH/antagonistas & inhibidores , VIH-1/efectos de los fármacos , Complejo SIDA Demencia/enzimología , Complejo SIDA Demencia/patología , Complejo SIDA Demencia/prevención & control , Complejo SIDA Demencia/virología , Amidohidrolasas/antagonistas & inhibidores , Fármacos Anti-VIH/uso terapéutico , Ácidos Araquidónicos/fisiología , Benzamidas/farmacología , Benzamidas/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Moduladores de Receptores de Cannabinoides/agonistas , Moduladores de Receptores de Cannabinoides/fisiología , Carbamatos/farmacología , Carbamatos/uso terapéutico , Línea Celular , Técnicas de Cocultivo , Endocannabinoides , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Endotelio Vascular/fisiología , Glicéridos/fisiología , Proteína gp120 de Envoltorio del VIH/fisiología , VIH-1/fisiología , Humanos , Microcirculación/efectos de los fármacos , Microcirculación/fisiología , Receptor Cannabinoide CB1/fisiología
7.
Mol Cell Biol ; 26(22): 8371-84, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16982692

RESUMEN

The actin-based cytoskeleton is essential for the generation and maintenance of cell polarity, cellular motility, and the formation of neural cell processes. MRP2 is an actin-binding protein of the kelch-related protein family. While MRP2 has been shown to be expressed specifically in brain, its function is still unknown. Here, we report that in neuronal growth factor (NGF)-induced PC12 cells, MRP2 was expressed along the neurite processes and colocalized with Talin at the growth cones. MRP2 mRNA and protein levels were up-regulated in PC12 cells following NGF stimulation. Moreover, treatment of PC12 cells with interfering RNAs for MRP2 and glycogen synthase kinase 3beta (GSK3beta) resulted in the inhibition of neurite outgrowth. A significant decrease in MRP2 expression levels was observed following GSK3beta inhibition, which was correlated with the inhibited neurite outgrowth, while GSK3beta overexpression was found to increase MRP2 expression levels. MRP2 interacted with GSK3beta through its NH2 terminus containing the BTB domain, and these molecules colocalized along neurite processes and growth cones in differentiated PC12 cells and rat primary hippocampal neurons. Additionally, increased associations of MRP2 with GSK3beta and MRP2 with actin were observed in the NGF-treated PC12 cells. Thus, this study provides, for the first time, insights into the involvement of MRP2 in neurite outgrowth, which occurs in a GSK3beta-dependent manner.


Asunto(s)
Glucógeno Sintasa Quinasa 3/fisiología , Proteínas de Microfilamentos/fisiología , Factor de Crecimiento Nervioso/farmacología , Neuritas/fisiología , Neuronas/fisiología , Actinas/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Neuritas/efectos de los fármacos , Neuritas/enzimología , Neuronas/enzimología , Neuronas/metabolismo , Células PC12 , ARN Interferente Pequeño/fisiología , Ratas , Ratas Sprague-Dawley , Receptor trkA/metabolismo , Transducción de Señal , Transfección
8.
Cancer Res ; 67(18): 8596-604, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875699

RESUMEN

The transcription factor NF-E2-related factor 2 (Nrf2) translocates into the nucleus and activates phase II genes encoding detoxification enzymes and antioxidant proteins, resulting in the protection of cells from oxidative insults. However, the involvement of Nrf2-mediated oxidative stress responses in breast cancer cells is largely unknown. Notably, during our study of the Nrf2 pathway in breast cancer cells, we observed that the nuclear matrix protein NRP/B was expressed and colocalized with Nrf2 in these cells, suggesting that NRP/B is involved in Nrf2-mediated oxidative stress responses. The expression level of NRP/B was variable in different breast cancer cells and breast cancer tissues, and was found to be localized in the nucleus. NRP/B expression was increased after exposure to the oxidative stress agent, hydrogen peroxide (H(2)O(2)), particularly in the highly aggressive MDA-MB-231 breast cancer cells. Association of NRP/B with Nrf2 in vitro and in vivo was observed in MDA-MB-231 breast cancer cells, and this association was up-regulated upon exposure to H(2)O(2), but not to sodium nitroprusside, SIN-1, and DETA-NO. NRP/B also enhanced Nrf2-mediated NAD(P)H:quinine oxidoreductase 1 promoter activity. Thus, this study reveals that NRP/B enhances oxidative stress responses in breast cancer cells via the Nrf2 pathway, identifying a novel role of nuclear matrix protein(s) in oxidative stress responses.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Microfilamentos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neuropéptidos/metabolismo , Proteínas Nucleares/metabolismo , Animales , Neoplasias de la Mama/genética , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Proteínas de Microfilamentos/biosíntesis , Proteínas de Microfilamentos/genética , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/genética , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Donantes de Óxido Nítrico/farmacología , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Estrés Oxidativo , Transfección , Regulación hacia Arriba
9.
Biochim Biophys Acta ; 1768(9): 2049-59, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17555706

RESUMEN

Cannabinoids are compounds that can modulate neuronal functions and immune responses via their activity at the CB(1) receptor. We used (2)H NMR order parameters and relaxation rate determination to delineate the behavior of magnetically aligned phospholipid bilayers in the presence of several structurally distinct cannabinoid ligands. THC (Delta(9)-Tetrahydrocannabinol) and WIN-55,212-2 were found to lower the phase transition temperature of the DMPC and to destabilize their acyl chains leading to a lower average S(CD) ( approximately 0.13), while methanandamide and CP-55,940 exhibited unusual properties within the lipid bilayer resulting in a greater average S(CD) ( approximately 0.14) at the top of the phospholipid upper chain. The CB(1) antagonist AM281 had average S(CD) values that were higher than the pure DMPC lipids, indicating a stabilization of the lipid bilayer. R(1Z) versus |S(CD)|(2) plots indicated that the membrane fluidity is increased in the presence of THC and WIN-55,212-2. The interaction of CP-55,940 with a variety of zwitterionic and charged membranes was also assessed. The unusual effect of CP-55,940 was present only in bicelles composed of DMPC. These studies strongly suggest that cannabinoid action on the membrane depends upon membrane composition as well as the structure of the cannabinoid ligands.


Asunto(s)
Cannabinoides/química , Cannabinoides/farmacología , Dimiristoilfosfatidilcolina/química , Membrana Dobles de Lípidos/química , Liposomas/química , Fluidez de la Membrana/efectos de los fármacos , Hidrógeno , Cinética , Espectroscopía de Resonancia Magnética , Conformación Molecular , Transición de Fase
10.
Cell Signal ; 19(2): 289-300, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16945503

RESUMEN

The protein tyrosine kinase RAFTK, also termed Pyk2, is a member of the focal adhesion kinase (FAK) subfamily. In this report, we show the role of RAFTK in neuroendocrine PC12 cells upon epidermal growth factor (EGF) stimulation. Following EGF treatment, we observed that RAFTK was tyrosine-phosphorylated in a time- and dose-dependent manner, while FAK was constitutively phosphorylated and primarily regulated by cell adhesion. Moreover, we found that RAFTK associated with the phosphorylated EGF receptor (EGFR) upon EGF stimulation. RAFTK phosphorylation was mediated primarily through PLCgamma-IP3-Ca(2+) signaling and partially through PI3-Kinase. Furthermore, overexpression of PRNK, a specific dominant-negative construct of RAFTK, was sufficient to block EGF-induced cell spreading and movement. Paxillin, a key modulator of the actin cytoskeleton and an RAFTK substrate, was also phosphorylated following EGF treatment. EGF induced a dynamic reorganization of RAFTK and paxillin at neuronal adhesion sites, with the specific localization of paxillin at the inner juxtaposition of RAFTK. Additionally, we observed that RAFTK associated with the scaffold protein c-Cbl and mediated its phosphorylation. Our data demonstrate that while FAK mediated cell adhesion, RAFTK was localized at the cytoplasm where it mediated inside-out signaling through intracellular Ca(2+), thus leading to cell spreading and movement upon EGF stimulation.


Asunto(s)
Citoplasma/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Quinasa 2 de Adhesión Focal/metabolismo , Animales , Señalización del Calcio , Adhesión Celular , Movimiento Celular , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 2 de Adhesión Focal/fisiología , Modelos Biológicos , Células PC12 , Paxillin/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Ratas , Ubiquitina/metabolismo
11.
Cancer Res ; 66(11): 5757-62, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16740714

RESUMEN

Amplification of the HER-2/neu (ErbB2) gene is observed in approximately 30% of human breast cancers, correlating with a poor clinical prognosis. Src kinases are also involved in the etiology of breast cancer, and their activation was suggested to be necessary for Neu-induced oncogenesis. To address whether Src activity is essential for Neu-mediated tumorigenesis, we used a physiologic inhibitor of Src kinase activity, the Csk homologous kinase (CHK), expressed as a mammary tissue-specific transgene. Our data, using a physiologic inhibitor of Src activity (CHK), showed that blocking of Neu-induced Src activity without altering Src expression levels had no significant effects on Neu-mediated mammary tumorigenesis in vivo. This contradicts the current paradigm that activation of Src kinases is essential for Neu-induced oncogenesis. This study is the first to distinguish between the kinase-dependent and kinase-independent actions of Src and shows that its kinase-dependent properties are not requisite for Neu-induced tumorigenesis.


Asunto(s)
Glándulas Mamarias Animales/enzimología , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Familia-src Quinasas/metabolismo , Animales , Femenino , Genes erbB-2/fisiología , Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , Virus del Tumor Mamario del Ratón/genética , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Fosforilación , Familia-src Quinasas/antagonistas & inhibidores
13.
PLoS Med ; 4(6): e186, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17550303

RESUMEN

BACKGROUND: While vascular endothelial growth factor (VEGF) expression in breast tumors has been correlated with a poor outcome in the pathogenesis of breast cancer, the expression, localization, and function of VEGF receptors VEGFR1 (also known as FLT1) and VEGFR2 (also known as KDR or FLK1), as well as neuropilin 1 (NRP1), in breast cancer are controversial. METHODS AND FINDINGS: We investigated the expression and function of VEGF and VEGF receptors in breast cancer cells. We observed that VEGFR1 expression was abundant, VEGFR2 expression was low, and NRP1 expression was variable. MDA-MB-231 and MCF-7 breast cancer cells, transfected with antisense VEGF cDNA or with siVEGF (VEGF-targeted small interfering RNA), showed a significant reduction in VEGF expression and increased apoptosis as compared to the control cells. Additionally, specifically targeted knockdown of VEGFR1 expression by siRNA (siVEGFR1) significantly decreased the survival of breast cancer cells through down-regulation of protein kinase B (AKT) phosphorylation, while targeted knockdown of VEGFR2 or NRP1 expression had no effect on the survival of these cancer cells. Since a VEGFR1-specific ligand, placenta growth factor (PGF), did not, as expected, inhibit the breast cancer cell apoptosis induced by siVEGF, and since VEGFR1 antibody also had no effects on the survival of these cells, we examined VEGFR1 localization. VEGFR1 was predominantly expressed internally in MDA-MB-231 and MCF-7 breast cancer cells. Specifically, VEGFR1 was found to be colocalized with lamin A/C and was expressed mainly in the nuclear envelope in breast cancer cell lines and primary breast cancer tumors. Breast cancer cells treated with siVEGFR1 showed significantly decreased VEGFR1 expression levels and a lack of VEGFR1 expression in the nuclear envelope. CONCLUSIONS: This study provides, to our knowledge for the first time, evidence of a unique survival system in breast cancer cells by which VEGF can act as an internal autocrine (intracrine) survival factor through its binding to VEGFR1. These results may lead to an improved strategy for tumor therapy based on the inhibition of angiogenesis.


Asunto(s)
Apoptosis/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis/genética , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunoprecipitación , Lamina Tipo A/metabolismo , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Desnudos , Mutación , Membrana Nuclear/metabolismo , Fosforilación , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología
14.
Cell Signal ; 18(7): 1063-71, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16199135

RESUMEN

The phospholipid lysophosphatidic acid (LPA) is a normal constituent of serum that functions as a lipid growth factor and intracellular signaling molecule. In this report, we have investigated the signaling mechanism and function of the tyrosine kinase RAFTK/Pyk2 in LPA-induced cell migration. Analysis of tyrosine phosphorylation upon LPA stimulation in neuroendocrine PC12 cells revealed 6 major tyrosine-phosphorylated proteins with estimated sizes of 180, 120, 115, 68, 44, and 42 kDa. These proteins were identified as epidermal growth factor receptor (EGFR), focal adhesion kinase, RAFTK/Pyk2, paxillin, Erk 1, and Erk 2, respectively. Using specific pharmacological inhibitors, we found that the tyrosine phosphorylation of RAFTK/Pyk2 was intracellular Ca2+-dependent, but not EGFR-dependent, during LPA stimulation of these cells. Moreover, the cytoskeletal and signal scaffolding protein, paxillin, associated with and was regulated by RAFTK/Pyk2 in a Ca2+-dependent manner. Characterization of LPA receptors showed that LPA1 (Edg2) and LPA2 (Edg4) are major receptors for LPA, while LPA3 receptor (Edg7) expression was limited. Upon using the LPA1/LPA3 receptor-specific antagonist VPC 32179, we observed that inhibition of the LPA1/LPA3 receptors had no effect on the LPA-induced phosphorylation of RAFTK, strongly suggesting that the LPA2 receptor is a key mediator of RAFTK phosphorylation. Furthermore, LPA induced PC12 cell migration, which was subsequently blocked by the dominant-negative form of FAK, FRNK. Expression of a dominant-negative form of the small GTPase Ras also blocked LPA-induced cell migration and RAFTK phosphorylation. Taken together, these results indicate that RAFTK is a key signaling molecule that mediates LPA-induced PC12 cell migration in a Ras-dependent manner.


Asunto(s)
Quinasa 2 de Adhesión Focal/fisiología , Lisofosfolípidos/fisiología , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Calcio/metabolismo , Movimiento Celular , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Quinasa 2 de Adhesión Focal/metabolismo , Lisofosfolípidos/farmacología , Células PC12 , Paxillin/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Ratas , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Tirosina/metabolismo
15.
Cell Signal ; 18(9): 1439-46, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16413169

RESUMEN

Dynamin-1 is a GTP-hydrolyzing protein and a key element in the clathrin-mediated endocytosis of secretory granules and neurovesicles at the plasma membrane. The unique receptor-like protein tyrosine phosphatase, PTP-NP/Phogrin/IAR/IA-2, is associated with neuroendocrine secretory granules and is highly expressed in the brain. Here, we show by confocal microscopy and biochemical studies that PTP-NP rapidly associates with Dynamin-1 in a depolarization-dependent manner and regulates Dynamin-1 GTPase activity upon KCl depolarization of rat primary hippocampal neurons. Depolarization of primary neurons induced direct association of PTP-NP with Dynamin-1 within 30 s. This association resulted in significant inhibition of Dynamin-1 GTPase activity (approximately 75% inhibition). Mutation within the phosphatase domain of PTP-NP (PTP-NP(D947A)) abolished the direct interaction of PTP-NP with Dynamin-1 and failed to inhibit Dynamin-1 GTPase activity. To further confirm the endogenous interaction of Dynamin-1 with wild-type PTP-NP, Dynamin-1 was purified biochemically from rat brain and its interaction with purified PTP-NP was analyzed. Highly purified Dynamin-1 specifically associated with wild-type PTP-NP and not with mutated PTP-NP, resulting in significant inhibition (approximately 70%) of Dynamin-1 GTPase activity. This is the first report to suggest a novel function of this unique receptor-type tyrosine phosphatase as a potential regulator of Dynamin-1 GTPase activity upon neuronal depolarization.


Asunto(s)
Autoantígenos/metabolismo , Dinamina I , Potenciales de la Membrana/fisiología , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Alanina/metabolismo , Animales , Autoantígenos/genética , Dinamina I/antagonistas & inhibidores , Dinamina I/metabolismo , Endocitosis/fisiología , Hipocampo/citología , Proteínas de la Membrana/genética , Neuronas/citología , Proteínas Tirosina Fosfatasas/genética , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores
16.
Cancer Res ; 65(7): 2840-5, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15805285

RESUMEN

Using microarray gene analysis, we found that carboxyl-terminal Src kinase homologous kinase (CHK) regulated the expression of the chemokine receptor, CXCR4. Northern blot and fluorescence-activated cell-sorting analyses showed that CHK down-regulated CXCR4 mRNA and protein levels, respectively. Mutated CHK, which contains a mutation within the ATP binding site of CHK, failed to inhibit CXCR4 expression, thus suggesting that CHK kinase activity is involved in the regulation of CXCR4. Results from gel shift analysis indicated that CHK regulates CXCR4 transcriptional activity by altering YY1 binding to the CXCR4 promoter. Whereas CHK had no significant effects on the expression of YY1, c-Myc, Max, and other YY1-binding proteins, CHK was found to modulate the YY1/c-Myc association. Furthermore, CHK inhibited CXCR4-positive breast cancer cell migration. Taken together, these studies show a novel mechanism by which CHK down-regulates CXCR4 through the YY1 transcription factor, leading to decreased CXCR4-mediated breast cancer cell motility and migration.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular/fisiología , Quimiocinas CXC/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas pp60(c-src)/fisiología , Receptores CXCR4/biosíntesis , Factores de Transcripción/fisiología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Línea Celular Tumoral , Quimiocina CXCL12 , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Factores de Unión al ADN Específico de las Células Eritroides , Humanos , Regiones Promotoras Genéticas , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/biosíntesis , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Receptores CXCR4/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Factor de Transcripción YY1
17.
Oncogene ; 24(14): 2398-409, 2005 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-15735724

RESUMEN

Mayven is a member of the kelch-related superfamily of proteins, characterized by a series of 'kelch' repeats at their carboxyl terminus and a BTB/POZ domain at their NH2-terminus. Little is known about the role of Mayven in cancer. Here, we report that Mayven expression was abundant and diffuse in primary human epithelial breast tumor cells as compared to normal breast epithelial cells, where Mayven was detected in the normal breast layer of the mammary ducts. Overexpression of Mayven resulted in an induction of c-Jun protein levels, as well as increased AP-1 (activating protein 1) transcriptional activity in MCF-7 and T47D breast cancer cells through its BTB/POZ domain. Furthermore, Mayven activated c-Jun N-terminal kinase in breast cancer cells. Mayven, through its BTB/POZ domain, induced cyclin D1 expression and cyclin D1 promoter activity and promoted cell cycle progression from the G1 to S phase. MCF-7 cells transduced with the recombinant retroviral sense Mayven (pMIG-W-Mayven) showed significant induction of c-Jun and cyclin D1 mRNA expression and activities as compared to the retroviral vector alone, while MCF-7 cells transduced by the recombinant retroviral antisense Mayven (pMIG-W-Mayven-AS) demonstrated a significant decrease in c-Jun and cyclin D1 expression and activities. Given the crucial functions of cyclin D1 and AP-1 signaling in oncogenesis, our results strongly suggest that overexpression of Mayven may promote tumor growth through c-Jun and cyclin D1.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ciclina D1/metabolismo , Proteínas de Microfilamentos/fisiología , Proteínas del Tejido Nervioso/fisiología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Fase G1 , Humanos , Inmunohistoquímica , Microscopía Fluorescente , Fase S
18.
Oncogene ; 23(35): 5890-900, 2004 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-15208678

RESUMEN

Nearly all brain tumors develop following the progressive accumulation of genetic alterations of oncogenes and tumor suppressor genes (such as p53 and retinoblastoma protein). Furthermore, aberrations in the nuclear matrix often contribute to genomic instabilities and the development of cancer. We have previously shown that nuclear-restricted protein/brain (NRP/B), a member of the BTB/Kelch repeat family, is a nuclear matrix protein normally expressed in neurons but not in astrocytes, and that it is an early and specific marker of neurons during the development of the central nervous system. Here, we show aberrant expression of NRP/B in human brain tissues. NRP/B is expressed in the cytoplasm of human brain tumor cells (glioblastoma, GBM) arising from astrocytes. NRP/B mutations (13 mutations in the Kelch domains, two in the intervening sequence (IVS) domain and two in the BTB domain) were detected in brain tumor cell lines (A-172, CCF-STTG1, SK-N-SH and U87-MG) and in primary human malignant GBM tissues (eight samples). More importantly, we found that NRP/B mutants, but not wild-type (wt) NRP/B, increased the activation of ERK and consequently promoted cell proliferation, attenuated caspase activation and suppressed the cellular apoptosis induced by the stressful stimulus cisplatin (10 microM). These events were observed to occur via a p53-mediated pathway. In addition, while wt NRP/B was associated with actin, mutations in the Kelch domains of NRP/B led to its reduced binding affinity to actin. Thus, alterations and gene mutations within the NRP/B gene may contribute to brain tumorigenesis by promoting cell proliferation, suppressing apoptosis and by affecting nuclear cytoskeleton dynamics.


Asunto(s)
Neoplasias Encefálicas/genética , Proteínas de Microfilamentos/genética , Mutación , Neuropéptidos/genética , Proteínas Nucleares/genética , Secuencias de Aminoácidos , Apoptosis , Encéfalo/metabolismo , Neoplasias Encefálicas/etiología , División Celular , Línea Celular Tumoral , Núcleo Celular/química , Humanos , Proteínas de Microfilamentos/análisis , Proteínas de Microfilamentos/química , Neuropéptidos/análisis , Neuropéptidos/química , Proteínas Nucleares/análisis , Proteínas Nucleares/química , Secuencias Repetitivas de Aminoácido , Proteína p53 Supresora de Tumor/fisiología
19.
Oncogene ; 21(50): 7730-9, 2002 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-12400015

RESUMEN

Mutational inactivation of BRCA1 confers increased risk for breast cancer. However, the underlying basis for the breast tissue-restricted, tumor-suppressive properties of BRCA1 remains poorly defined. Here, we show that BRCA1 and the estrogen receptor alpha (ER-alpha) modulated vascular endothelial growth factor (VEGF) gene transcription and secretion in breast cancer cells. ER-alpha interacted in vitro and in vivo with BRCA1, and this interaction was mediated by the AF-2 domain of ER-alpha and two domains of BRCA1, the amino-acid residues 1-306 and 428-683. Endogenous interaction of ER-alpha with BRCA1 was observed in normal MCF-10A breast epithelial cells and in breast cancer cells (MCF-7 and T47D), and this interaction was significantly reduced in the presence of estrogen. Furthermore, ER-alpha induced activation of VEGF gene transcription, using human VEGF promoter-luciferase reporter constructs. The AF-2 domain of ER-alpha was also shown to induce VEGF gene transcription activation similar to that obtained with the full-length ER-alpha. However, in the presence of BRCA1, VEGF gene transcription activation and VEGF protein secretion were significantly inhibited in a dose-dependent manner. The BRCA1 domain of 1-683 amino acid residues was required for this inhibition of VEGF gene transcription activation. Three mutated forms of BRCA1 (A1708E, M1775R and Y1853X), that have been identified in familial breast cancers, failed to associate with ER-alpha and to suppress VEGF promoter activity and VEGF protein secretion. Overexpression of wild-type BRCA1 in HCC-1937 breast cancer cells that lack endogenous functional BRCA1 significantly reduced VEGF secretion in these cells. These results demonstrate a novel pathogenic mechanism whereby mutations in BRCA1, via their interaction with ER-alpha, could promote tumorigenesis through the hormonal regulation of mammary epithelial cell proliferation and impaired VEGF function, which may lead to cancer growth and angiogenesis.


Asunto(s)
Proteína BRCA1/metabolismo , Neoplasias de la Mama/metabolismo , Factores de Crecimiento Endotelial/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Linfocinas/genética , Neoplasias Hormono-Dependientes/metabolismo , Receptores de Estrógenos/metabolismo , Proteína BRCA1/genética , Mama/citología , Neoplasias de la Mama/genética , Células Cultivadas , Regulación hacia Abajo , Factores de Crecimiento Endotelial/metabolismo , Células Epiteliales , Receptor alfa de Estrógeno , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Linfocinas/metabolismo , Mutación , Neoplasias Hormono-Dependientes/genética , Estructura Terciaria de Proteína , Receptores de Estrógenos/genética , Transcripción Genética , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
20.
Mol Cancer Res ; 2(6): 327-38, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15235108

RESUMEN

In this study, we have characterized the signaling pathways mediated by CXCR4 in breast cancer cells and its role in breast cancer cell invasion and migration. Stromal cell-derived factor 1alpha (SDF-1alpha; CXCL12) stimulation of breast cancer cells resulted in phosphoinositide 3-kinase (PI-3K) activation, AKT phosphorylation, and activation of the FKHRL1 transcription factor. In addition, SDF-1alpha induced activation of the focal adhesion kinase (FAK) as well as the migration of breast cancer cells. Expression of SDF-1alpha, the ligand of CXCR4, was about 2-fold higher in microdissected human breast epithelial cancer cells as compared with normal epithelial cells. Immunohistochemical analysis indicated that SDF-1alpha expression is consistently higher in primary breast tumor cells than in normal breast epithelial cells. Furthermore, SDF-1alpha induced blood vessel instability, through increased vascular permeability, resulting in the penetration of breast tumor cells through the human brain microvascular endothelial cells (HBMEC). Notably, the migration of breast cancer cells was inhibited by the PI-3K inhibitor, Wortmannin, and the Ca(2+) inhibitor BAPTA/AM, indicating that transendothelial breast cancer cell migration induced by SDF-1alpha is mediated by activation of the PI-3K/AKT pathway and Ca(2+)-mediated signaling. Blockade of the CXCR4/SDF1 signaling pathway with anti-CXCR4 antibody also decreased transendothelial breast cancer cell migration as well as vascular permeability. This study focuses on novel interactions between highly relevant signaling pathways in breast cancer cells and brain microvascular endothelial cells and may provide insights into the molecular mechanisms of CXCR4/SDF-1alpha-mediated breast cancer metastasis to the brain.


Asunto(s)
Encéfalo/irrigación sanguínea , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular/efectos de los fármacos , Quimiocinas CXC/farmacología , Células Endoteliales/citología , Receptores CXCR4/metabolismo , Calcio/metabolismo , Línea Celular Tumoral , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Proteínas de Unión al ADN/metabolismo , Activación Enzimática/efectos de los fármacos , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead , Humanos , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA