Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cells ; 13(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38727288

RESUMEN

Glioblastoma (GBM) is a devastating brain cancer for which new effective therapies are urgently needed. GBM, after an initial response to current treatment regimens, develops therapeutic resistance, leading to rapid patient demise. Cancer cells exhibit an inherent elevation of endoplasmic reticulum (ER) stress due to uncontrolled growth and an unfavorable microenvironment, including hypoxia and nutrient deprivation. Cancer cells utilize the unfolded protein response (UPR) to maintain ER homeostasis, and failure of this response promotes cell death. In this study, as integrins are upregulated in cancer, we have evaluated the therapeutic potential of individually targeting all αß1 integrin subunits using RNA interference. We found that GBM cells are uniquely susceptible to silencing of integrin α3. Knockdown of α3-induced proapoptotic markers such as PARP cleavage and caspase 3 and 8 activation. Remarkably, we discovered a non-canonical function for α3 in mediating the maturation of integrin ß1. In its absence, generation of full length ß1 was reduced, immature ß1 accumulated, and the cells underwent elevated ER stress with upregulation of death receptor 5 (DR5) expression. Targeting α3 sensitized TRAIL-resistant GBM cancer cells to TRAIL-mediated apoptosis and led to growth inhibition. Our findings offer key new insights into integrin α3's role in GBM survival via the regulation of ER homeostasis and its value as a therapeutic target.


Asunto(s)
Estrés del Retículo Endoplásmico , Glioblastoma , Integrina alfa3 , Integrina beta1 , Ligando Inductor de Apoptosis Relacionado con TNF , Humanos , Apoptosis/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Glioblastoma/patología , Glioblastoma/metabolismo , Glioblastoma/genética , Integrina alfa3/metabolismo , Integrina alfa3/genética , Integrina beta1/metabolismo , Integrina beta1/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
2.
Bioorg Med Chem Lett ; 21(18): 5528-32, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21831638

RESUMEN

Solid tumors generally grow under hypoxic conditions, a pathophysiological change, which activates the expression of genes responsible for malignant, aggressive, and treatment-refractory properties. Hypoxia inducible factor (HIF) is the chief transcription factor regulating hypoxia-driven gene expression. Therefore, the HIF pathway has become a critical target for cancer therapeutics development. We screened a privileged library of about 10,000 natural-product-like compounds using a cell-based assay for HIF-dependent transcriptional activity and identified several arylsulfonamide HIF pathway inhibitors. Among these compounds, the most potent ones showed an IC(50) of ∼0.5 µM in the hypoxia-responsive element (HRE)-luciferase reporter system. Further studies are needed to fully elucidate the mechanism of action of this class of compounds and their structure-activity relationship.


Asunto(s)
Sulfonamidas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Fosfatasa Alcalina/antagonistas & inhibidores , Fosfatasa Alcalina/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Factor 1 Inducible por Hipoxia , Estructura Molecular , Bibliotecas de Moléculas Pequeñas , Estereoisomerismo , Relación Estructura-Actividad , Sulfonamidas/química , Factores de Transcripción/metabolismo
3.
Oncogene ; 40(33): 5182-5191, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34218269

RESUMEN

Uveal melanoma (UM) is the most prevalent primary intraocular malignancy in adults, and patients that develop metastases (~50%) survive <1 year, highlighting the urgent need for new therapies. TCGA has recently revealed that a hypoxia gene signature is associated with poor UM patient prognosis. Here we show that expression of hypoxia-regulated collagen prolyl-4-hydroxylase genes P4HA1 and P4HA2 is significantly upregulated in UM patients with metastatic disease and correlates with poor prognosis, suggesting these enzymes might be key tumor drivers. We targeted hypoxia-induced expression of P4HA1/2 in UM with KCN1, a hypoxia inducible factor-1 (HIF-1) pathway inhibitor and found potent inhibition of primary and metastatic disease and extension of animal survival, without overt side effects. At the molecular level, KCN1 antagonized hypoxia-induced expression of P4HA1 and P4HA2, which regulate collagen maturation and deposition in the extracellular matrix. The treatment decreased prolyl hydroxylation, induced proteolytic cleavage and rendered a disordered structure to collagen VI, the main collagen produced by UM, and reduced UM cell invasion. Together, these data demonstrate that extracellular collagen matrix formation can be targeted in UM by inhibiting hypoxia-induced P4HA1 and P4HA2 expression, warranting further development of this strategy in patients with uveal melanoma.


Asunto(s)
Prolina Dioxigenasas del Factor Inducible por Hipoxia , Matriz Extracelular , Humanos , Hidroxilación , Melanoma , Activación Transcripcional , Regulación hacia Arriba , Neoplasias de la Úvea
4.
Oncogene ; 39(5): 1165, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31676871

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Oncogene ; 39(5): 1041-1048, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31582835

RESUMEN

Medulloblastoma (MB) is a malignant pediatric brain tumor for which new therapies are urgently needed. We demonstrate that treatment with EPZ-6438 (Tazemetostat), an enhancer of zeste homolog 2 (EZH2) inhibitor approved for clinical trials, blocks MB cell growth in vitro and in vivo, and prolongs survival in orthotopic xenograft models. We show that the therapeutic effect is dependent on epigenetic reactivation of adhesion G-protein-coupled receptor B1 (BAI1/ADGRB1), a tumor suppressor that controls p53 stability by blocking Mdm2. Histone 3 trimethylated on lysine 27 (H3K27me3), a marker of silent chromatin conformation is present at the ADGRB1 promoter, and inhibition of EZH2, the catalytic component of the Polycomb Repressive complex 2 (PRC2) that methylates H3K27, switches the gene into an active chromatin status and reactivates BAI1 expression. Mechanistically, targeting EZH2 promotes transition from H3K27me3 to H3K27ac at the promoter, recruits the C/EBPß (CREB-binding protein) and CBP transcription factors and activates ADGRB1 gene transcription. Taken together, our results identify key molecular players that regulate ADGRB1 gene expression in MB, demonstrate that reactivation of BAI1 expression underlies EPZ-6438 antitumorigenic action, and provide preclinical proof-of-principle evidence for targeting EZH2 in patients with MB.


Asunto(s)
Proteínas Angiogénicas/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética , Meduloblastoma/patología , Receptores Acoplados a Proteínas G/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Angiogénicas/deficiencia , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Transformación Celular Neoplásica , Silenciador del Gen , Histonas/metabolismo , Humanos , Metilación , Fragmentos de Péptidos/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Sialoglicoproteínas/metabolismo
6.
Clin Cancer Res ; 26(11): 2711-2724, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31969339

RESUMEN

PURPOSE: Exploitation of altered glycosylation in cancer is a major goal for the design of new cancer therapy. Here, we designed a novel secreted chimeric signal peptide-Galectin-3 conjugate (sGal-3) and investigated its ability to induce cancer-specific cell death by targeting aberrantly N-glycosylated cell surface receptors on cancer cells. EXPERIMENTAL DESIGN: sGal-3 was genetically engineered from Gal-3 by extending its N-terminus with a noncleavable signal peptide from tissue plasminogen activator. sGal-3 killing ability was tested on normal and tumor cells in vitro and its antitumor activity was evaluated in subcutaneous lung cancer and orthotopic malignant glioma models. The mechanism of killing was investigated through assays detecting sGal-3 interaction with specific glycans on the surface of tumor cells and the elicited downstream proapoptotic signaling. RESULTS: We found sGal-3 preferentially binds to ß1 integrin on the surface of tumor cells due to aberrant N-glycosylation resulting from cancer-associated upregulation of several glycosyltransferases. This interaction induces potent cancer-specific death by triggering an oncoglycan-ß1/calpain/caspase-9 proapoptotic signaling cascade. sGal-3 could reduce the growth of subcutaneous lung cancers and malignant gliomas in brain, leading to increased animal survival. CONCLUSIONS: We demonstrate that sGal-3 kills aberrantly glycosylated tumor cells and antagonizes tumor growth through a novel integrin ß1-dependent cell-extrinsic apoptotic pathway. These findings provide proof-of-principle that aberrant N-oncoglycans represent valid cancer targets and support further translation of the chimeric sGal-3 peptide conjugate for cancer therapy.


Asunto(s)
Apoptosis , Proteínas Sanguíneas/metabolismo , Galectinas/metabolismo , Glioma/tratamiento farmacológico , Integrina beta1/metabolismo , Fragmentos de Péptidos/farmacología , Señales de Clasificación de Proteína , Animales , Proteínas Sanguíneas/genética , Proliferación Celular , Femenino , Galectinas/genética , Glioma/metabolismo , Glioma/patología , Glicosilación , Humanos , Integrina beta1/genética , Ratones , Ratones Desnudos , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Clin Cancer Res ; 25(7): 2206-2218, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30563937

RESUMEN

PURPOSE: Uveal melanoma (UM) is the most prevalent and lethal intraocular malignancy in adults. Here, we examined the importance of hypoxia in UM growth and tested the antitumor effects of arylsulfonamide 64B, an inhibitor of the hypoxia-induced factor (HIF) pathway in animal models of UM and investigated the related mechanisms. EXPERIMENTAL DESIGN: UM cells were implanted in the uvea of mice eyes and mice systemically treated with 64B. Drug effect on primary eye tumor growth, circulating tumor cells, metastasis formation in liver, and survival were examined. 64B effects on UM cell growth, invasion and hypoxia-induced expression of C-X-C chemokine receptor type 4 (CXCR4) and mesenchymal-epithelial transition factor (c-Met) were measured. Luciferase reporter assays, chromatin immunoprecipitation, co-immunoprecipitation, and cellular thermal shift assays were used to determine how 64B interferes with the HIF transcriptional complex. RESULTS: Systemic administration of 64B had potent antitumor effects against UM in several orthotopic mouse models, suppressing UM growth in the eye (∼70% reduction) and spontaneous liver metastasis (∼50% reduction), and extending mice survival (P < 0.001) while being well tolerated. 64B inhibited hypoxia-induced expression of CXCR4 and c-Met, 2 key drivers of tumor invasion and metastasis. 64B disrupted the HIF-1 complex by interfering with HIF-1α binding to p300/CBP co-factors, thus reducing p300 recruitment to the MET and CXCR4 gene promoters. 64B could thermostabilize p300, supporting direct 64B binding to p300. CONCLUSIONS: Our preclinical efficacy studies support the further optimization of the 64B chemical scaffold toward a clinical candidate for the treatment of UM.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Melanoma/etiología , Melanoma/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Receptores CXCR4/genética , Sulfonamidas/farmacología , Neoplasias de la Úvea/etiología , Neoplasias de la Úvea/metabolismo , Animales , Biomarcadores de Tumor , Biopsia , Línea Celular Tumoral , Modelos Animales de Enfermedad , Proteína p300 Asociada a E1A/metabolismo , Humanos , Neoplasias Hepáticas/secundario , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Pronóstico , Unión Proteica , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores CXCR4/metabolismo , Sulfonamidas/química , Neoplasias de la Úvea/tratamiento farmacológico , Neoplasias de la Úvea/patología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Cell ; 33(6): 1004-1016.e5, 2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29894688

RESUMEN

Adhesion G protein-coupled receptors (ADGRs) encompass 33 human transmembrane proteins with long N termini involved in cell-cell and cell-matrix interactions. We show the ADGRB1 gene, which encodes Brain-specific angiogenesis inhibitor 1 (BAI1), is epigenetically silenced in medulloblastomas (MBs) through a methyl-CpG binding protein MBD2-dependent mechanism. Knockout of Adgrb1 in mice augments proliferation of cerebellar granule neuron precursors, and leads to accelerated tumor growth in the Ptch1+/- transgenic MB mouse model. BAI1 prevents Mdm2-mediated p53 polyubiquitination, and its loss substantially reduces p53 levels. Reactivation of BAI1/p53 signaling axis by a brain-permeable MBD2 pathway inhibitor suppresses MB growth in vivo. Altogether, our data define BAI1's physiological role in tumorigenesis and directly couple an ADGR to cancer formation.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Neoplasias Cerebelosas/metabolismo , Meduloblastoma/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Angiogénicas/genética , Animales , Línea Celular Tumoral , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/genética , Células HCT116 , Humanos , Estimación de Kaplan-Meier , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Proteínas Proto-Oncogénicas c-mdm2/genética , Interferencia de ARN , Receptores Acoplados a Proteínas G , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Oncogene ; 24(22): 3632-42, 2005 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-15782143

RESUMEN

Brain angiogenesis inhibitor 1 (BAI1) is a transmembrane protein with unknown function expressed primarily in normal but not tumoral brain. The finding of thrombospondin type 1 repeats in its extracellular domain suggested an antiangiogenic function, but the mechanisms by which a transmembrane receptor could inhibit angiogenesis remained unexplained. Here we demonstrate that BAI1 is proteolytically cleaved at a conserved G-protein-coupled receptor proteolytic cleavage site (GPS), releasing its 120 kDa extracellular domain. We named this secreted fragment Vasculostatin as it inhibited migration of endothelial cells in vitro and dramatically reduced in vivo angiogenesis. Both constitutive and doxycycline-induced expression of Vasculostatin elicited dose-dependent suppression of tumor growth and vascular density in mice, implicating Vasculostatin in the regulation of vascular homeostasis and tumor prevention. Generation of a soluble antiangiogenic factor by cleavage of a pre-existing transmembrane protein represents a novel mechanism for regulating vascular homeostasis and preventing tumorigenesis. Modulation of this cleavage or delivery of Vasculostatin may constitute novel treatment modalities for cancer and other diseases of aberrant angiogenesis, especially in the brain.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Proteínas Angiogénicas/farmacología , Antineoplásicos/farmacología , Fragmentos de Péptidos/farmacología , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/genética , Proteínas Angiogénicas/genética , Animales , Western Blotting , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Femenino , Glioma/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Neovascularización Patológica/metabolismo , Fragmentos de Péptidos/genética , Receptores Acoplados a Proteínas G
10.
Clin Cancer Res ; 22(24): 6256-6265, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27340277

RESUMEN

PURPOSE: Elevation in D-2-Hydroxyglutarate (D-2HG) has recently emerged as a mandatory byproduct of mutated Isocitrate Dehydrogenase (IDH) genes 1 and 2 in glioma patients. The goal of the present study was to demonstrate the feasibility of detection of elevated levels of D-2HG in the cerebrospinal fluid (CSF) of glioma patients that carry point substitutions in the IDH gene. EXPERIMENTAL DESIGN: We developed a mass spectrometry (MS)-based platform to detect and quantify the D- and L-forms of 2HG in the CSF of glioma patients. Three independent cohorts of patients were analyzed, comprising a total of 176 samples derived from 84 patients. The levels of D- and L-2HG were used to stratify patients into IDH wild-type or IDH-mutated groups using an empirically obtained threshold of 0.69 µmol/L. RESULTS: Using this platform, a greater than 17-fold mean increase in D-2HG was observed in the CSF of patients with IDH mutant versus wild-type gliomas. The means for the D-2HG levels in CSF were 0.427 µmol/L in wild-type and 7.439 µmol/L in mutant groups. The C statistic for the receiver operator curve was 0.938, with 84% sensitivity, 90% specificity, and 89% accuracy to detect D-2HG. The levels of D- and L-2HG in CSF from wild-type patients varied by location of CSF draw (cisternal > ventricular > lumbar). CONCLUSIONS: Our findings demonstrate that the CSF of patients harboring IDH mutant gliomas contain increased levels of D-2HG, which can be reliably detected with a MS-based platform. Clin Cancer Res; 22(24); 6256-65. ©2016 AACR.


Asunto(s)
Neoplasias Encefálicas/genética , Líquido Cefalorraquídeo/metabolismo , Glioma/genética , Glutaratos/líquido cefalorraquídeo , Isocitrato Deshidrogenasa/genética , Mutación/genética , Adulto , Anciano , Anciano de 80 o más Años , Encéfalo/metabolismo , Neoplasias Encefálicas/líquido cefalorraquídeo , Femenino , Glioma/líquido cefalorraquídeo , Humanos , Masculino , Espectrometría de Masas/métodos , Persona de Mediana Edad , Adulto Joven
11.
Clin Cancer Res ; 18(24): 6623-33, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22923450

RESUMEN

PURPOSE: The hypoxia-inducible factor-1 (HIF-1) plays a critical role in tumor adaptation to hypoxia, and its elevated expression correlates with poor prognosis and treatment failure in patients with cancer. In this study, we determined whether 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide, KCN1, the lead inhibitor in a novel class of arylsulfonamide inhibitors of the HIF-1 pathway, had antitumorigenic properties in vivo and further defined its mechanism of action. EXPERIMENTAL DESIGN: We studied the inhibitory effect of systemic KCN1 delivery on the growth of human brain tumors in mice. To define mechanisms of KCN1 anti-HIF activities, we examined its influence on the assembly of a functional HIF-1α/HIF-1ß/p300 transcription complex. RESULTS: KCN1 specifically inhibited HIF reporter gene activity in several glioma cell lines at the nanomolar level. KCN1 also downregulated transcription of endogenous HIF-1 target genes, such as VEGF, Glut-1, and carbonic anhydrase 9, in a hypoxia-responsive element (HRE)-dependent manner. KCN1 potently inhibited the growth of subcutaneous malignant glioma tumor xenografts with minimal adverse effects on the host. It also induced a temporary survival benefit in an intracranial model of glioma but had no effect in a model of melanoma metastasis to the brain. Mechanistically, KCN1 did not downregulate the levels of HIF-1α or other components of the HIF transcriptional complex; rather, it antagonized hypoxia-inducible transcription by disrupting the interaction of HIF-1α with transcriptional coactivators p300/CBP. CONCLUSIONS: Our results suggest that the new HIF pathway inhibitor KCN1 has antitumor activity in mouse models, supporting its further translation for the treatment of human tumors displaying hypoxia or HIF overexpression.


Asunto(s)
Antineoplásicos/farmacología , Benzopiranos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Proteína de Unión a CREB/antagonistas & inhibidores , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Glioma/tratamiento farmacológico , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Sulfonamidas/farmacología , Animales , Neoplasias Encefálicas/patología , Proteína de Unión a CREB/metabolismo , Línea Celular Tumoral , Proteína p300 Asociada a E1A/metabolismo , Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Glioma/patología , Humanos , Concentración 50 Inhibidora , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Unión Proteica/efectos de los fármacos , Elementos de Respuesta , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Med Chem ; 54(24): 8471-89, 2011 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-22032632

RESUMEN

Hypoxia, a reduction in partial oxygen pressure, is a salient property of solid tumors. Hypoxia drives malignant progression and metastasis in tumors and participates in tumor resistance to radio- and chemotherapies. Hypoxia activates the hypoxia-inducible factor (HIF) family of transcription factors, which induce target genes that regulate adaptive biological processes such as anaerobic metabolism, cell motility, and angiogenesis. Clinical evidence has demonstrated that expression of HIF-1 is strongly associated with poor patient prognosis and activation of HIF-1 contributes to malignant behavior and therapeutic resistance. Consequently, HIF-1 has become an important therapeutic target for inhibition by small molecules. Herein, we describe the design and synthesis of small molecules that inhibit the HIF-1 signaling pathway. Many of these compounds exhibit inhibitory activity in the nanomolar range. Separate mechanistic studies indicate that these inhibitors do not alter HIF-1 levels but interfere with the ability of HIF-1α/HIF-1ß to interact with cofactors p300/CBP to form an active transcriptional complex.


Asunto(s)
Antineoplásicos/síntesis química , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Antineoplásicos/química , Antineoplásicos/farmacología , Translocador Nuclear del Receptor de Aril Hidrocarburo/antagonistas & inhibidores , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Benzofuranos/síntesis química , Benzofuranos/química , Benzofuranos/farmacología , Benzopiranos/síntesis química , Benzopiranos/química , Benzopiranos/farmacología , Proteína de Unión a CREB/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Piridinas/síntesis química , Piridinas/química , Piridinas/farmacología , Quinolinas/síntesis química , Quinolinas/química , Quinolinas/farmacología , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Factores de Transcripción p300-CBP/metabolismo
13.
Cancer Res ; 69(3): 1212-20, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19176395

RESUMEN

Angiogenesis is a critical physiologic process that is appropriated during tumorigenesis. Little is known about how this process is specifically regulated in the brain. Brain angiogenesis inhibitor-1 (BAI1) is a brain-predominant seven-transmembrane protein that contains five antiangiogenic thrombospondin type-1 repeats (TSR). We recently showed that BAI1 is cleaved at a conserved proteolytic cleavage site releasing a soluble, 120 kDa antiangiogenic factor called vasculostatin (Vstat120). Vstat120 has been shown to inhibit in vitro angiogenesis and suppress subcutaneous tumor growth. Here, we examine its effect on the intracranial growth of malignant gliomas and further study its antitumor mechanism. First, we show that expression of Vstat120 strongly suppresses the intracranial growth of malignant gliomas, even in the presence of the strong proangiogenic stimulus mediated by the oncoprotein epidermal growth factor receptor variant III (EGFRvIII). This tumor-suppressive effect is accompanied by a decrease in tumor vascular density, suggesting a potent antiangiogenic effect in the brain. Second, and consistent with this interpretation, we find that treatment with Vstat120 reduces the migration of cultured microvascular endothelial cells in vitro and inhibits corneal angiogenesis in vivo. Third, we show that these antivascular effects critically depend on the presence of the cell surface receptor CD36 on endothelial cells in vitro and in vivo, supporting the role of Vstat120 TSRs in mediating these effects. These results advance the understanding of brain-specific angiogenic regulation, and suggest that Vstat120 has therapeutic potential in the treatment of brain tumors and other intracerebral vasculopathies.


Asunto(s)
Proteínas Angiogénicas/biosíntesis , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/terapia , Antígenos CD36/metabolismo , Glioblastoma/irrigación sanguínea , Glioblastoma/terapia , Fragmentos de Péptidos/biosíntesis , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Movimiento Celular/fisiología , Neovascularización de la Córnea/tratamiento farmacológico , ADN Complementario/administración & dosificación , ADN Complementario/genética , Células Endoteliales/patología , Glioblastoma/genética , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/terapia , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Ratas , Receptores Acoplados a Proteínas G , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cancer Res ; 68(17): 6953-62, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18757409

RESUMEN

BRCA1-associated protein-1 (BAP1), a deubiquitinating enzyme of unknown cellular function, is mutated in breast and lung cancers. In this study, we have shown for the first time that BAP1 has tumor suppressor activity in vivo by showing that BAP1 can suppress tumorigenicity of lung cancer cells in athymic nude mice. We show that BAP1 fulfills another criterion of a genuine tumor suppressor because cancer-associated BAP1 mutants are deficient in deubiquitinating activity. We show for the first time that one of the two predicted nuclear targeting motifs is required for nuclear localization of BAP1 and that a truncation mutant found in a lung cancer cell line results in BAP1 that fails to localize to the nucleus. Furthermore, we show that deubiquitinating activity and nuclear localization are both required for BAP1-mediated tumor suppression in nude mice. We show that BAP1 exerts its tumor suppressor functions by affecting the cell cycle, speeding the progression through the G(1)-S checkpoint, and inducing cell death via a process that has characteristics of both apoptosis and necrosis. Surprisingly, BAP1-mediated growth suppression is independent of wild-type BRCA1. Because deubiquitinating enzymes are components of the ubiquitin proteasome system, this pathway has emerged as an important target for anticancer drugs. The identification of the deubiquitinating enzyme BAP1 as a tumor suppressor may lead to further understanding of how the ubiquitin proteasome system contributes to cancer and aid in the identification of new targets for cancer therapy.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas Supresoras de Tumor/fisiología , Ubiquitina Tiolesterasa/fisiología , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Animales , Ciclo Celular , División Celular , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/química , Ubiquitina Tiolesterasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA