Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Nat Med ; 12(8): 933-8, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16862154

RESUMEN

CD82, also known as KAI1, was recently identified as a prostate cancer metastasis suppressor gene on human chromosome 11p1.2 (ref. 1). The product of CD82 is KAI1, a 40- to 75-kDa tetraspanin cell-surface protein also known as the leukocyte cell-surface marker CD82 (refs. 1,2). Downregulation of KAI1 has been found to be clinically associated with metastatic progression in a variety of cancers, whereas overexpression of CD82 specifically suppresses tumor metastasis in various animal models. To define the mechanism of action of KAI1, we used a yeast two-hybrid screen and identified an endothelial cell-surface protein, DARC (also known as gp-Fy), as an interacting partner of KAI1. Our results indicate that the cancer cells expressing KAI1 attach to vascular endothelial cells through direct interaction between KAI1 and DARC, and that this interaction leads to inhibition of tumor cell proliferation and induction of senescence by modulating the expression of TBX2 and p21. Furthermore, the metastasis-suppression activity of KAI1 was significantly compromised in DARC knockout mice, whereas KAI1 completely abrogated pulmonary metastasis in wild-type and heterozygous littermates. These results provide direct evidence that DARC is essential for the function of CD82 as a suppressor of metastasis.


Asunto(s)
Sistema del Grupo Sanguíneo Duffy/metabolismo , Endotelio Vascular/metabolismo , Proteína Kangai-1/metabolismo , Neoplasias Pulmonares/patología , Glicoproteínas de Membrana/metabolismo , Metástasis de la Neoplasia/prevención & control , Receptores de Superficie Celular/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Senescencia Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Sistema del Grupo Sanguíneo Duffy/química , Femenino , Heterocigoto , Humanos , Proteína Kangai-1/química , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Ratas , Receptores de Superficie Celular/química , Proteínas de Dominio T Box/metabolismo
2.
Breast Cancer Res Treat ; 128(1): 57-68, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20665104

RESUMEN

We previously isolated cacalol as a free radical-scavenging compound from Cacalia delphiniifolia which is a traditional Asian herbal plant and is believed to have medicinal effects on cancer. In this report, we demonstrated that cacalol has strong anti-proliferation effect on breast cancer cells and induces apoptosis by activating a pro-apoptotic pathway. We also found that a combination of cacalol and other chemotherapeutic drugs (Taxol and cyclophosphamide) synergistically induced apoptosis and partially overcame chemo-resistance. To further gain a mechanistic insight, we tested a potential inhibitory effect of cacalol on fatty acid synthase gene (FAS) in breast cancer cells, and found that cacalol significantly modulated the expression of the FAS gene, which resulted in apoptosis through activation of DAPK2 and caspase 3. We have also shown that cacalol significantly suppressed the Akt-sterol regulatory element-binding proteins (SREBP) signaling pathway and concomitant transcriptional activation of FAS. In a xenograft model of nude mouse, when cacalol was administered intraperitoneally, tumor growth was significantly suppressed. Importantly, oral administration of cacalol before implanting tumors showed significant preventive effect on tumor growth in the same animal model. Furthermore, the treatment of mice with a combination of low dose of Taxol and cacalol significantly suppressed the tumor growth. Taken together, our results indicate that cacalol induces apoptosis in breast cancer cells and impairs mammary tumor growth in vivo by blocking the expression of the FAS gene through modulation of Akt-SREBP pathway, suggesting that cacalol has potential utility as a chemopreventive and chemotherapeutic agent for breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ácido Graso Sintasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sesquiterpenos/farmacología , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Antineoplásicos/efectos adversos , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias de la Mama , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Caspasa 3/metabolismo , Línea Celular Tumoral , Ciclofosfamida/farmacología , Proteínas Quinasas Asociadas a Muerte Celular , Sinergismo Farmacológico , Activación Enzimática , Ácido Graso Sintasas/genética , Femenino , Genes Reporteros , Humanos , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Ratones , Ratones Desnudos , Paclitaxel/farmacología , Regiones Promotoras Genéticas , Sesquiterpenos/efectos adversos , Transducción de Señal , Transcripción Genética , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Biochim Biophys Acta ; 1786(2): 87-104, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18692117

RESUMEN

It is well recognized that the majority of cancer related deaths is caused by metastatic diseases. Therefore, there is an urgent need for the development of therapeutic intervention specifically targeted to the metastatic process. In the last decade, significant progress has been made in this research field, and many new concepts have emerged that shed light on the molecular mechanism of metastasis cascade which is often portrayed as a succession of six distinct steps; localized invasion, intravasation, translocation, extravasation, micrometastasis and colonization. Successful metastasis is dependent on the balance and complex interplay of both the metastasis promoters and suppressors in each step. Therefore, the basic strategy of our interventions is aimed at either blocking the promoters or potentiating the suppressors in this disease process. Toward this goal, various kinds of antibodies and small molecules have been designed. These include agents that block the ligand-recepter interaction of metastasis promoters (HGF/c-Met), antagonize the metastasis-promoting enzymes (AMF, uPA and MMP) and inhibit the transcriptional activity of metastasis promoter (beta-Catenin). On the other hand, the intriguing roles of metastasis suppressors and their signal pathways have been extensively studied and various attempts have been made to potentiate these factors. Small molecules have been developed to restore the expression or mimic the function of metastasis-suppressor genes such as NM23, E-cadherin, Kiss-1, MKK4 and NDRG1, and some of them are under clinical trials. This review summarizes our current understanding of the molecular pathway of tumor metastasis and discusses strategies and recent development of anti-metastatic drugs.


Asunto(s)
Antineoplásicos/uso terapéutico , Genes Supresores de Tumor/fisiología , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/genética , Neoplasias/tratamiento farmacológico , Carcinógenos/antagonistas & inhibidores , Sistemas de Liberación de Medicamentos , Humanos , Modelos Biológicos , Neoplasias/genética , Neoplasias/patología , Transducción de Señal/efectos de la radiación , Proteínas Supresoras de Tumor/uso terapéutico
4.
Cancer Res ; 67(4): 1411-4, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17308076

RESUMEN

Tumor metastases suppressor protein KAI1/CD82 is capable of blocking the tumor metastases without affecting the primary tumor formation, and its expression is significantly down-regulated in many types of human cancers. However, the exact molecular mechanism of the suppressor function of KAI1 remains elusive. Evidence from our laboratory supports a model in which tumor cells dislodge from the primary tumor and intravasate into the blood or lymphatic vessels followed by attachment to the endothelial cell surface whereby KAI1 interacts with the Duffy antigen receptor for chemokines (DARC) protein. This interaction transmits a senescent signal to cancer cells expressing KAI1, whereas cells that lost KAI1 expression can proliferate, potentially giving rise to metastases. Our model of the mechanism of action of KAI1 shows that metastasis suppressor activity can be dependent on interaction with host tissue and explains how KAI1 suppresses metastasis without affecting primary tumor formation. Taken together, in vitro and in vivo studies identify the KAI1-DARC interaction as a potential target for cancer therapy.


Asunto(s)
Sistema del Grupo Sanguíneo Duffy/metabolismo , Proteína Kangai-1/metabolismo , Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Células Endoteliales/metabolismo , Humanos , Metástasis de la Neoplasia , Neoplasias/patología
5.
Cancer Res ; 66(11): 5934-40, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16740734

RESUMEN

Fatty acid synthase (FAS) has been found to be overexpressed in a wide range of epithelial tumors, including breast cancer. Pharmacologic inhibitors of FAS cause apoptosis of breast cancer cells and result in decreased tumor size in vivo. However, how the inhibition of FAS induces apoptosis in tumor cells remains largely unknown. To understand the apoptotic pathway resulting from direct inhibition of FAS, we treated breast tumor cells with or without FAS small interfering RNA (siRNA) followed by a microarray analysis. Our results indicated that the proapoptotic genes BNIP3, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), and death-associated protein kinase 2 (DAPK2) were significantly up-regulated on direct inhibition of the FAS gene. We also found that the knockdown of FAS expression significantly increased ceramide level in the tumor cells, and this increase was abrogated by acetyl-CoA carboxylase inhibitor. In addition, carnitine palmitoyltransferase-1 (CPT-1) inhibitor up-regulated the ceramide and BNIP3 levels in these cells, whereas treatment of tumor cells with FAS siRNA in the presence of a ceramide synthase inhibitor abrogated the up-regulation of BNIP3 and inhibited apoptosis. Furthermore, we found that treatment of cells with BNIP3 siRNA significantly counteracted the effect of FAS siRNA-mediated apoptosis. Consistent with these results, a significant inverse correlation was observed in the expression of FAS and BNIP3 in clinical samples of human breast cancer. Collectively, our results indicate that inhibition of FAS in breast cancer cells causes accumulation of malonyl-CoA, which leads to inhibition of CPT-1 and up-regulation of ceramide and induction of the proapoptotic genes BNIP3, TRAIL, and DAPK2, resulting in apoptosis.


Asunto(s)
Apoptosis/fisiología , Neoplasias de la Mama/enzimología , Ácido Graso Sintasas/antagonistas & inhibidores , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Línea Celular Tumoral , Proteínas Quinasas Asociadas a Muerte Celular , Ácido Graso Sintasas/genética , Humanos , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , Ligando Inductor de Apoptosis Relacionado con TNF , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba
6.
Cancer Res ; 66(24): 11983-90, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17178897

RESUMEN

The tumor metastasis suppressor gene Drg-1 has been shown to suppress metastasis without affecting tumorigenicity in immunodeficient mouse models of prostate and colon cancer. Expression of Drg-1 has also been found to have a significant inverse correlation with metastasis or invasiveness in various types of human cancer. However, how Drg-1 exerts its metastasis suppressor function remains unknown. In the present study, to elucidate the mechanism of action of the Drg-1 gene, we did a microarray analysis and found that induction of Drg-1 significantly inhibited the expression of activating transcription factor (ATF) 3, a member of the ATF/cyclic AMP-responsive element binding protein family of transcription factors. We also showed that Drg-1 attenuated the endogenous level of ATF3 mRNA and protein in prostate cancer cells, whereas Drg-1 small interfering RNA up-regulated the ATF3 expression. Furthermore, Drg-1 suppressed the promoter activity of the ATF3 gene, indicating that Drg-1 regulates ATF3 expression at the transcriptional level. Our immunohistochemical analysis on prostate cancer specimens revealed that nuclear expression of ATF3 was inversely correlated to Drg-1 expression and positively correlated to metastases. Consistently, we have found that ATF3 overexpression promoted invasiveness of prostate tumor cells in vitro, whereas Drg-1 suppressed the invasive ability of these cells. More importantly, overexpression of ATF3 in prostate cancer cells significantly enhanced spontaneous lung metastasis of these cells without affecting primary tumorigenicity in a severe combined immunodeficient mouse model. Taken together, our results strongly suggest that Drg-1 suppresses metastasis of prostate tumor cells, at least in part, by inhibiting the invasive ability of the cells via down-regulation of the expression of the ATF3 gene.


Asunto(s)
Factor de Transcripción Activador 3/genética , Proteínas de Unión al GTP/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/genética , Línea Celular Tumoral , Humanos , Masculino , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/prevención & control , Plásmidos , Neoplasias de la Próstata/patología , Mapeo Restrictivo , Transfección
7.
Oncogene ; 24(34): 5389-95, 2005 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-15897909

RESUMEN

Fatty acid synthase (FAS), a key enzyme of the fatty acid biosynthetic pathway, has been shown to be overexpressed in various types of human cancer and is, therefore, considered to be an attractive target for anticancer therapy. However, the exact mechanism of overexpression of the FAS gene in tumor cells is not well understood. In this report, we demonstrate that the expression of the tumor suppressor gene PTEN has a significant inverse correlation with FAS expression in the case of prostate cancer in the clinical setting, and inhibition of the PTEN gene leads to the overexpression of FAS in vitro. We also found that the combination of the expression status of these two genes is a better prognostic marker than either gene alone. Furthermore, our results indicate that the specific inhibition of FAS gene by siRNA leads to apoptosis of prostate tumor cells, and inhibition of PI 3-kinase pathway synergizes with FAS siRNA to enhance tumor cell death. These results provide a strong rationale for exploring the therapeutic use of an inhibitor of the PTEN signaling pathway in conjunction with the FAS siRNA to inhibit prostate tumor growth.


Asunto(s)
Apoptosis , Ácido Graso Sintasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas/metabolismo , Monoéster Fosfórico Hidrolasas/biosíntesis , Neoplasias de la Próstata/genética , Proteínas Supresoras de Tumor/biosíntesis , Anciano , Anciano de 80 o más Años , Ácido Graso Sintasas/biosíntesis , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Fosfohidrolasa PTEN , Pronóstico , Neoplasias de la Próstata/patología , Interferencia de ARN , Transducción de Señal , Análisis de Supervivencia
8.
Front Biosci ; 11: 2845-60, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16720356

RESUMEN

Despite significant improvement in surgical techniques and chemotherapies, none of the current medical technologies "cure" metastatic disease, and the patients who have acquired metastatic cancer inevitably die from disseminated disease. Thus, there is a need for developing novel therapeutic approaches which can directly target metastatic tumor cells. However, advances in understanding the molecular mechanism of tumor metastases have lagged behind other developments in the cancer field. Tumor metastasis involves complex array of steps with each step requiring a coordination of the actions of many positive and negative factors. A number of tumor metastasis suppressors have been identified which suppress the formation of tumor metastasis without affecting the growth rate of the primary tumor. Such discoveries offer new approaches for curtailing tumor metastasis. This review summarizes our current understanding on these genes and their potential role in the progression of tumor metastases.


Asunto(s)
Neoplasias de la Mama/patología , Genes Supresores de Tumor , Metástasis de la Neoplasia/genética , Neoplasias de la Próstata/patología , Neoplasias de la Mama/genética , Femenino , Genes Relacionados con las Neoplasias , Humanos , Masculino , Neoplasias de la Próstata/genética
9.
Cancer Res ; 62(5): 1246-50, 2002 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11888884

RESUMEN

To search for p53 target genes throughout the human genome, we applied a cDNA microarray system using adenovirus-mediated transfer of p53 into p53-deficient U373MG (glioblastoma) cells. In this manner, we detected dozens of genes that appeared to be regulated by wild-type p53. We describe here characterization of one such gene, termed CABC1 [chaperone-activity of bc1 complex in Schizosaccharomyces pombe (ABC1)-like], which encodes a 647-amino acid peptide with significant sequence similarity to activity of bc1 complex (ABC1) in Arabidopsis thaliana and S. pombe. The CABC1 product was located in mitochondria, and colony-formation assays with cancer cell lines indicated its ability to suppress cell growth. Inhibition of CABC1 expression by transfection with antisense oligonucleotide significantly reduced the apoptotic response induced by wild-type p53. These results suggest that CABC1 may play an important role in mediating p53-inducible apoptosis through the mitochondrial pathway.


Asunto(s)
Proteínas Fúngicas/química , Proteínas de la Membrana/genética , Mitocondrias/química , Proteínas Mitocondriales/genética , Proteínas de Saccharomyces cerevisiae , Proteínas de Schizosaccharomyces pombe/genética , Secuencia de Aminoácidos , Animales , Apoptosis , Células COS , Daño del ADN , Humanos , Mitocondrias/fisiología , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Transcripción Genética , Proteína p53 Supresora de Tumor/fisiología
10.
Cancer Res ; 64(17): 5963-72, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15342375

RESUMEN

To characterize the molecular feature in prostate carcinogenesis and the putative transition from prostatic intraepithelial neoplasia (PIN) to invasive prostate cancer (PC), we analyzed gene-expression profiles of 20 PCs and 10 high-grade PINs with a cDNA microarray representing 23,040 genes. Considering the histological heterogeneity of PCs and the minimal nature of PIN lesions, we applied laser microbeam microdissection to purify populations of PC and PIN cells, and then compared their expression profiles with those of corresponding normal prostatic epithelium also purified by laser microbeam microdissection. A hierarchical clustering analysis separated the PC group from the PIN group, except for three tumors that were morphologically defined as one very-high-grade PIN and two low-grade PCs, suggesting that PINs and PCs share some molecular features and supporting the hypothesis of PIN-to-PC transition. On the basis of this hypothesis, we identified 21 up-regulated genes and 63 down-regulated genes commonly in PINs and PCs compared with normal epithelium, which were considered to be involved in the presumably early stage of prostatic carcinogenesis. They included AMACR, OR51E2, RODH, and SMS. Furthermore, we identified 41 up-regulated genes and 98 down-regulated genes in the transition from PINs to PCs; those altered genes, such as POV1, CDKN2C, EPHA4, APOD, FASN, ITGB2, LAMB2, PLAU, and TIMP1, included elements that are likely to be involved in cell adhesion or the motility of invasive PC cells. The down-regulation of EPHA4 by small interfering RNA in PC cells lead to attenuation of PC cell viability. These data provide clues to the molecular mechanisms underlying prostatic carcinogenesis, and suggest candidate genes the products of which might serve as molecular targets for the prevention and treatment of PC.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasia Intraepitelial Prostática/genética , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , División Celular/genética , Transformación Celular Neoplásica/patología , Análisis por Conglomerados , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño/genética , Receptor EphA4/antagonistas & inhibidores , Receptor EphA4/biosíntesis , Receptor EphA4/genética
11.
Oncogene ; 21(18): 2914-8, 2002 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-11973653

RESUMEN

Interferon regulatory factors (IRFs) regulate transcription of interferon genes through DNA sequence-specific binding to these targets. Using a differential display method for examining gene expression in p53-defective cells infected with adenovirus containing wild-type p53, we found that expression of interferon regulatory factor 5 (IRF-5) mRNA was increased in the presence of exogenous p53. An electrophoretic mobility-shift assay showed that a potential p53 binding site (p53BS) detected in exon 2 of the IRF-5 gene could in fact bind to p53 protein. Moreover, a heterologous reporter assay revealed that the p53BS possessed p53-dependent transcriptional activity. Expression of IRF-5 was induced in p53+/+ cells (MCF7 and NHDF), but not inp53-/- cells (H1299) when DNA was damaged by gamma-irradiation, UV-radiation, or adriamycin treatment in a wild-type p53-dependent manner. These results suggest that IRF-5 is a novel p53-target, and that it might mediate the p53-dependent immune response.


Asunto(s)
Proteínas de Unión al ADN/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , División Celular , Humanos , Factores Reguladores del Interferón , Datos de Secuencia Molecular , Activación Transcripcional , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
12.
Oncogene ; 23(32): 5551-7, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15077175

RESUMEN

In an earlier study designed to investigate molecular mechanisms of carcinogenesis in synovial sarcomas (SSs), we applied a cDNA microarray to detect human genes with significantly increased expression in SS cells. Among the genes selected in this way, we identified a novel transcript, subsequently designated PDZK4 (PDZ domain-containing 4), that was specifically upregulated in all of the 13 SS cases we examined. On Northern blots of normal human tissues, the PDZK4 transcript was expressed only in fetal brain. Immunocytochemical staining of transfected COS7 cells showed that the PDZK4 gene product localized mainly under the plasma membrane. Treatment of human SS cells with small interfering RNA (siRNA) inhibited the expression of PDZK4 and resulted in the suppression of tumor-cell growth. Induction of exogenous PDZK4 expression promoted growth of T98G and COS7 cells in which no endogenous expression of PDZK4 was observed. Taken together, these findings strongly suggest that inappropriate expression of PDZK4 might play an important role in the proliferation of SS cells and that the gene might be a suitable molecular target for designing of novel drugs to treat SS patients.


Asunto(s)
Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Sarcoma Sinovial/metabolismo , Regulación hacia Arriba , Secuencia de Bases , Northern Blotting , Encéfalo/metabolismo , Cromosomas Humanos X , Silenciador del Gen/fisiología , Humanos , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma Sinovial/genética
13.
EMBO Mol Med ; 5(3): 384-96, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23495140

RESUMEN

Brain metastasis of breast cancer profoundly affects the cognitive and sensory functions as well as morbidity of patients, and the 1 year survival rate among these patients remains less than 20%. However, the pathological mechanism of brain metastasis is as yet poorly understood. In this report, we found that metastatic breast tumour cells in the brain highly expressed IL-1ß which then 'activated' surrounding astrocytes. This activation significantly augmented the expression of JAG1 in the astrocytes, and the direct interaction of the reactivated astrocytes and cancer stem-like cells (CSCs) significantly stimulated Notch signalling in CSCs. We also found that the activated Notch signalling in CSCs up-regulated HES5 followed by promoting self-renewal of CSCs. Furthermore, we have shown that the blood-brain barrier permeable Notch inhibitor, Compound E, can significantly suppress the brain metastasis in vivo. These results represent a novel paradigm for the understanding of how metastatic breast CSCs re-establish their niche for their self-renewal in a totally different microenvironment, which opens a new avenue to identify a novel and specific target for the brain metastatic disease.


Asunto(s)
Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Células Madre Neoplásicas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Antineoplásicos/farmacología , Astrocitos/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/prevención & control , Neoplasias de la Mama/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Comunicación Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células 3T3 NIH , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Interferencia de ARN , Ratas , Receptores Notch/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Serrate-Jagged , Transducción de Señal/efectos de los fármacos , Nicho de Células Madre , Factores de Tiempo , Transfección , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cancer Res ; 68(18): 7613-20, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18794150

RESUMEN

RhoC is a member of the Ras-homologous family of genes which have been implicated in tumorigenesis and tumor progression. However, the exact role of RhoC is controversial and is yet to be clarified. We have examined the effect of RhoC on prostate tumor cells and found that RhoC had no effect on cell proliferation in vitro or on tumor growth in mice. However, RhoC significantly enhanced the metastatic ability of the tumor cells in these animals, suggesting that RhoC affects only the metastasis but not the growth of prostate tumor cells. The results of our immunohistochemical analyses on tumor specimens from 63 patients with prostate cancer indicate that RhoC expression had no significant correlation with Gleason grade. However, the expression of RhoC showed significant positive correlation with both lymph node and distant metastasis, and it was inversely correlated with patient survival. We also found that RhoC significantly augmented the invasion and motility of prostate tumor cells by activating matrix metalloproteinases 2 and 9 (MMP2 and MMP9) in vitro. The results of our antibody array analysis for signal molecules revealed that RhoC significantly activated kinases including mitogen-activated protein kinase (MAPK), focal adhesion kinase (FAK), Akt, and Pyk2. Inhibition of Pyk2 kinase blocked the RhoC-dependent activation of FAK, MAPK, and Akt, followed by the suppression of MMP2 and MMP9. Inhibitors of both MAPK and Akt also significantly blocked the activities of these MMPs. Therefore, our results indicate that RhoC promotes tumor metastasis in prostate cancer by sequential activation of Pyk2, FAK, MAPK, and Akt followed by the up-regulation of MMP2 and MMP9, which results in the stimulation of invasiveness of tumor cells.


Asunto(s)
Quinasa 2 de Adhesión Focal/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Activación Enzimática , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Metástasis de la Neoplasia , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Ratas , Transducción de Señal , Regulación hacia Arriba , Proteínas de Unión al GTP rho/biosíntesis , Proteína rhoC de Unión a GTP
15.
Cancer Res ; 68(4): 1003-11, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18281474

RESUMEN

The fatty acid synthase (FAS) gene is significantly up-regulated in various types of cancers, and blocking the FAS expression results in apoptosis of tumor cells. Therefore, FAS is considered to be an attractive target for anticancer therapy. However, the molecular mechanism by which the FAS gene is up-regulated in tumor cells is poorly understood. We found that FAS was significantly up-regulated by hypoxia, which was also accompanied by reactive oxygen species (ROS) generation in human breast cancer cell lines. The FAS expression was also activated by H(2)O(2), whereas N-acetyl-L-cystein, a ROS inhibitor, suppressed the expression. We also found that the hypoxia significantly up-regulated sterol regulatory-element binding protein (SREBP)-1, the major transcriptional regulator of the FAS gene, via phosphorylation of Akt followed by activation of hypoxia-inducible factor 1 (HIF1). Moreover, our results of reporter assay and chromatin immunoprecipitation analysis indicate that SREBP-1 strongly bound to the SREBP binding site/E-box sequence on the FAS promoter under hypoxia. In our xenograft mouse model, FAS was strongly expressed in the hypoxic regions of the tumor. In addition, our results of immunohistochemical analysis for human breast tumor specimens indicate that the expressions of both FAS and SREBP-1 were colocalized with hypoxic regions in the tumors. Furthermore, we found that hypoxia-induced chemoresistance to cyclophosphamide was partially blocked by a combination of FAS inhibitor and cyclophosphamide. Taken together, our results indicate that FAS gene is up-regulated by hypoxia via activation of the Akt and HIF1 followed by the induction of the SREBP-1 gene, and that hypoxia-induced chemoresistance is partly due to the up-regulation of FAS.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Ácido Graso Sintasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Hipoxia de la Célula/genética , Línea Celular Tumoral , Ciclofosfamida/administración & dosificación , Ciclofosfamida/farmacología , Resistencia a Antineoplásicos , Activación Enzimática , Inhibidores Enzimáticos/administración & dosificación , Ácido Graso Sintasas/antagonistas & inhibidores , Ácido Graso Sintasas/biosíntesis , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Factor 1 Inducible por Hipoxia/biosíntesis , Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Desnudos , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
16.
Microvasc Res ; 74(2-3): 114-20, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17498748

RESUMEN

Metastasis is the primary cause of death in cancer patients. However, the molecular mechanism of the metastatic process is poorly understood because it involves multiple steps with a high degree of complexity. A critical step for successful establishment of secondary colonization is the hematogenous dissemination of malignant cells. During this process, the attachment of cancer cells to the endothelial cells on microvasculature is considered to be an essential step and many adhesion molecules as well as chemokines have been found to be involved in this process. This interaction of cancer-endothelial cell is considered not only to determine the physical site of metastasis, but also to provide the necessary anchorage to facilitate tumor cell extravasation. However, recent evidence indicates that this interaction also serves as a host defense mechanism and hinders the process of metastasis. The tumor metastases suppressor gene, KAI1, has been known to block metastatic process without affecting the primary tumor growth, and this protein has been found to be able to bind to the chemokine receptor, Duffy antigen receptor for chemokines (DARC), which is expressed on endothelial cells. Importantly, this interaction markedly induces senescence of tumor cells. This novel finding is not only significant in the context of molecular dissection of metastatic process but also in the therapeutic implication to develop drugs inhibiting metastasis.


Asunto(s)
Células Endoteliales/metabolismo , Genes Supresores de Tumor , Metástasis de la Neoplasia/genética , Neoplasias/metabolismo , Animales , Sistema del Grupo Sanguíneo Duffy/metabolismo , Genes Supresores de Tumor/fisiología , Humanos , Proteína Kangai-1/metabolismo , Modelos Biológicos , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/fisiopatología , Neoplasias/genética , Neoplasias/patología , Receptores de Superficie Celular/metabolismo
17.
Int J Cancer ; 118(10): 2441-7, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16380976

RESUMEN

Prostate cancer is a major cause of cancer death and morbidity in western countries. However, because of its intrinsic nature of chemoresistance, there is only limited systemic therapy available for the patients. Vitamin E (VE) has been under intensive study as a chemopreventive agent for various types of cancers. Preclinical studies suggest that vitamin E succinate (VES) is the most effective antitumor analogue of VE, yet there are scarce studies of VES in prostate cancer. In this study, we investigated the effects of VES on a panel of prostate cancer cells, and a xenograft model of prostate cancer. Our results indicate that VES significantly inhibited proliferation and induced apoptosis of prostate cancer cell lines in a dose and time dependent manner. The results of microarray analysis followed by real-time RT-PCR and inhibitor analyses indicated that the VES-induced apoptosis is mediated by caspase-4 in prostate tumor cells. In our animal model of prostate cancer in SCID mouse, daily injection of VES significantly suppressed tumor growth as well as lung metastases. These results suggest a potential therapeutic utility of VES for patients with prostate cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Próstata/patología , Vitamina E/análogos & derivados , Animales , Caspasas/biosíntesis , Caspasas/metabolismo , Caspasas Iniciadoras , Proliferación Celular/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tocoferoles , Trasplante Heterólogo , Células Tumorales Cultivadas , Vitamina E/farmacología
18.
Cancer Sci ; 97(11): 1211-6, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16965393

RESUMEN

To isolate novel diagnostic markers and drug targets for pancreatic ductal adenocarcinoma (PDAC), we previously performed expression profile analysis of PDAC cells using a genome-wide cDNA microarray combined with laser microdissection. Among dozens of up-regulated genes identified in PDAC cells, we herein focused on one tyrosine kinase receptor, Eph receptor A4 (EphA4), as a molecular target for PDAC therapy. Immunohistochemical analysis validated EphA4 overexpression in approximately half of the PDAC tissues. To investigate its biological function in PDAC cells, we knocked down EphA4 expression by siRNA, which drastically attenuated PDAC cell viability. In concordance with the siRNA experiment, PDAC-derivative cells that were designed to constitutively express exogenous EphA4 showed a more rapid growth rate than cells transfected with mock vector, suggesting a growth-promoting effect of EphA4 on PDAC cells. Furthermore, the expression analysis for ephrin ligand family members indicated the coexistence of ephrinA3 ligand in PDAC cells with EphA4 receptor, and knockdown of ephrinA3 by siRNA also attenuated PDAC cell viability. These results suggest that the EphA4-ephrinA3 pathway is likely to be a promising molecular target for pancreatic cancer therapy.


Asunto(s)
Proliferación Celular , Neoplasias Pancreáticas/metabolismo , Receptor EphA4/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Northern Blotting , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Receptor EphA4/antagonistas & inhibidores , Receptor EphA4/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA