Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Mol Cell ; 53(4): 534-48, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24486017

RESUMEN

Mitochondrial pyruvate dehydrogenase complex (PDC) is crucial for glucose homeostasis in mammalian cells. The current understanding of PDC regulation involves inhibitory serine phosphorylation of pyruvate dehydrogenase (PDH) by PDH kinase (PDK), whereas dephosphorylation of PDH by PDH phosphatase (PDP) activates PDC. Here, we report that lysine acetylation of PDHA1 and PDP1 is common in epidermal growth factor (EGF)-stimulated cells and diverse human cancer cells. K321 acetylation inhibits PDHA1 by recruiting PDK1, and K202 acetylation inhibits PDP1 by dissociating its substrate PDHA1, both of which are important in promoting glycolysis in cancer cells and consequent tumor growth. Moreover, we identified mitochondrial ACAT1 and SIRT3 as the upstream acetyltransferase and deacetylase, respectively, of PDHA1 and PDP1, while knockdown of ACAT1 attenuates tumor growth. Furthermore, Y381 phosphorylation of PDP1 dissociates SIRT3 and recruits ACAT1 to PDC. Together, hierarchical, distinct posttranslational modifications act in concert to control molecular composition of PDC and contribute to the Warburg effect.


Asunto(s)
Acetil-CoA C-Acetiltransferasa/metabolismo , Piruvato Deshidrogenasa (Lipoamida)-Fosfatasa/metabolismo , Piruvato Deshidrogenasa (Lipoamida)/metabolismo , Sirtuina 3/metabolismo , Tirosina/química , Animales , Línea Celular Tumoral , Proliferación Celular , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Lisina/química , Masculino , Ratones , Ratones Desnudos , Mitocondrias/metabolismo , Trasplante de Neoplasias , Neoplasias/metabolismo , Fosforilación
2.
Bioorg Med Chem Lett ; 27(8): 1731-1736, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28285917

RESUMEN

Many forms of solid tumor have a characteristic feature known as hypoxia, which describes a low or non-existent presence of oxygen in the cellular microenvironment. This decrease in oxygen causes activation of the hypoxia inducible factor (HIF) pathway, which activates the transcription of many genes that cause cell proliferation, metastasis, increased glycolysis and angiogenesis. Increased HIF expression has been linked with poor patient prognosis, increased malignancy, and therapeutic resistance. Previous work in our lab has identified 1 and 2 as inhibitors of the HIF pathway, specifically as disrupters of the p300-HIF-1α complex formation. A library of sulfonamide analogs has been designed and synthesized with the intent of examining the SAR of this series of compounds and improving potency and physicochemical properties as compared with lead compounds 1 and 2. At the end, we have achieved a thorough understanding of the structural features critical for future optimization work.


Asunto(s)
Benzopiranos/química , Benzopiranos/farmacología , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Proteína p300 Asociada a E1A/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Simulación del Acoplamiento Molecular , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
3.
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
4.
Bioorg Med Chem ; 20(14): 4590-7, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22682301

RESUMEN

We have discovered that 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide, a novel small molecule HIF-1 pathway inhibitor, can antagonize tumor growth in animal models of cancer, but the treatment necessitates its delivery in a formulation, due to poor water solubility (<15 µg/mL; pH 7.4), evidencing that the chemotype needs further exploration of its amenability to additional chemical modifications for ultimate optimization of function and pharmacology. As a first step towards this goal we investigated the structure-activity relationships of 15 lipophilic 2,2-dimethyl-2H-chromene based arylsulfonamide analogs of 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide to find out strategies of modification. A 3,4-dimethoxybenzenesulfonyl group in region 1 showed the strongest inhibition among five arylsulfonyl groups tested. The presence of propan-2-amine in region 2 conferred the strongest inhibitory effect of the compound on HIF-1 activated transcription in a reporter assay. These findings are important as they help define the structural motifs where the 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide can be chemically modified to improve its pharmacological properties towards development as a cancer therapeutic.


Asunto(s)
Antineoplásicos/química , Benzopiranos/química , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Sulfonamidas/química , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Línea Celular Tumoral , Genes Reporteros , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/farmacología , Transcripción Genética/efectos de los fármacos
5.
Mol Biol Cell ; 33(2): ar13, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34818063

RESUMEN

ELMODs are a family of three mammalian paralogues that display GTPase-activating protein (GAP) activity toward a uniquely broad array of ADP-ribosylation factor (ARF) family GTPases that includes ARF-like (ARL) proteins. ELMODs are ubiquitously expressed in mammalian tissues, highly conserved across eukaryotes, and ancient in origin, being present in the last eukaryotic common ancestor. We described functions of ELMOD2 in immortalized mouse embryonic fibroblasts (MEFs) in the regulation of cell division, microtubules, ciliogenesis, and mitochondrial fusion. Here, using similar strategies with the paralogues ELMOD1 and ELMOD3, we identify novel functions and locations of these cell regulators and compare them to those of ELMOD2, allowing the determination of functional redundancy among the family members. We found strong similarities in phenotypes resulting from deletion of either Elmod1 or Elmod3 and marked differences from those arising in Elmod2 deletion lines. Deletion of either Elmod1 or Elmod3 results in the decreased ability of cells to form primary cilia, loss of a subset of proteins from cilia, and accumulation of some ciliary proteins at the Golgi, predicted to result from compromised traffic from the Golgi to cilia. These phenotypes are reversed upon activating mutant expression of either ARL3 or ARL16, linking their roles to ELMOD1/3 actions.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Factores de Ribosilacion-ADP/metabolismo , Animales , Cilios/metabolismo , Proteínas del Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Proteínas Activadoras de GTPasa/fisiología , Aparato de Golgi/metabolismo , Ratones , Microtúbulos/metabolismo , Dinámicas Mitocondriales , Transducción de Señal/genética
6.
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
7.
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
8.
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
9.
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.

10.
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
11.
Cancer Res ; 67(14): 6872-81, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17638898

RESUMEN

There is a need for novel therapies targeting hypoxic cells in tumors. These cells are associated with tumor resistance to therapy and express hypoxia inducible factor-1 (HIF-1), a transcription factor that mediates metabolic adaptation to hypoxia and activates tumor angiogenesis. We previously developed an oncolytic adenovirus (HYPR-Ad) for the specific killing of hypoxic/HIF-active tumor cells, which we now armed with an interleukin-4 gene (HYPR-Ad-IL4). We designed HYPR-Ad-IL4 by cloning the Ad E1A viral replication and IL-4 genes under the regulation of a bidirectional hypoxia/HIF-responsive promoter. The IL-4 cytokine was chosen for its ability to induce a strong host antitumor immune response and its potential antiangiogenic activity. HYPR-Ad-IL4 induced hypoxia-dependent IL-4 expression, viral replication, and conditional cytolysis of hypoxic, but not normoxic cells. The treatment of established human tumor xenografts with HYPR-Ad-IL4 resulted in rapid and maintained tumor regression with the same potency as that of wild-type dl309-Ad. HYPR-Ad-IL4-treated tumors displayed extensive necrosis, fibrosis, and widespread viral replication. Additionally, these tumors contained a distinctive leukocyte infiltrate and prominent hypoxia. The use of an oncolytic Ad that locally delivers IL-4 to tumors is novel, and we expect that HYPR-Ad-IL4 will have broad therapeutic use for all solid tumors that have hypoxia or active HIF, regardless of tissue origin or genetic alterations.


Asunto(s)
Adenoviridae/metabolismo , Terapia Genética/métodos , Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Viroterapia Oncolítica/métodos , Virus Oncolíticos/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Clonación Molecular , Humanos , Hipoxia , Ratones , Modelos Genéticos , Trasplante de Neoplasias
12.
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
13.
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
15.
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
16.
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
17.
Clin Cancer Res ; 10(24): 8603-12, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15623644

RESUMEN

Hypoxia plays a critical role in driving tumor malignancy and is associated with poor patient survival in many human cancers. Novel therapies targeting hypoxic tumor cells are urgently needed, because these cells hinder tumor eradication. Here we demonstrate than an anticancer strategy based on intratumoral delivery of a novel type of oncolytic adenovirus targeting tumor hypoxia is therapeutically efficient and can augment standard chemotherapy. We used a conditionally replicative adenovirus (HYPR-Ad) to specifically kill hypoxic tumor cells. Viral infection and conditional replication occurred efficiently in hypoxic/hypoxia-inducible factor-active cells in culture and in vivo, prevented tumor formation, and reduced the growth of established tumors. Combining HYPR-Ad with chemotherapy effective against normoxic cells resulted in strongly enhanced antitumor efficacy. These studies demonstrate that targeting the hypoxic microenvironment of tumors rather than an intrinsic gene expression defect is a viable and novel antitumor therapeutic strategy that can be used in combination with existing treatment regimens. The replication and oncolytic potential of this virus was made dependent on hypoxic/hypoxia-inducible factor, a transcription factor activated in the tumor hypoxic microenvironment, broadening its therapeutic use to solid tumors of any genetic make-up or tissue of origin.


Asunto(s)
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Neoplasias Encefálicas/terapia , Hipoxia de la Célula/genética , Glioma/terapia , Replicación Viral/fisiología , Animales , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/virología , Carmustina/uso terapéutico , Terapia Combinada , Proteínas de Unión al ADN/metabolismo , Glioma/genética , Glioma/virología , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Ratones Desnudos , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Med Chem ; 55(15): 6738-50, 2012 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-22746274

RESUMEN

The hypoxia inducible factor (HIF) pathway is an attractive target for cancer, as it controls tumor adaptation to growth under hypoxia and mediates chemotherapy and radiation resistance. We previously discovered 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide as a novel, small-molecule HIF-1 pathway inhibitor in a high-throughput cell-based assay, but its in vivo delivery is hampered by poor aqueous solubility (0.009 µM in water; log P(7.4) = 3.7). Here we describe the synthesis of 12 N-alkyl-N-[(8-R-2,2-dimethyl-2H-chromen-6-yl)methyl]heteroarylsulfonamides, which were designed to possess optimal lipophilicities and aqueous solubilities by in silico calculations. Experimental log P(7.4) values of 8 of the 12 new analogs ranged from 1.2-3.1. Aqueous solubilities of three analogs were measured, among which the most soluble N-[(8-methoxy-2,2-dimethyl-2H-chromen-6-yl)methyl]-N-(propan-2-yl)pyridine-2-sulfonamide had an aqueous solubility of 80 µM, e.g., a solubility improvement of ∼9000-fold. The pharmacological optimization had limited impact on drug efficacy as the compounds retained IC(50) values at or below 5 µM in our HIF-dependent reporter assay.


Asunto(s)
Antineoplásicos/síntesis química , Benzopiranos/síntesis química , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Sulfonamidas/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Benzopiranos/química , Benzopiranos/farmacología , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Transducción de Señal , Solubilidad , Estereoisomerismo , Relación Estructura-Actividad , Sulfonamidas/química , Sulfonamidas/farmacología , Transcripción Genética/efectos de los fármacos
19.
ACS Med Chem Lett ; 3(8): 620-5, 2012 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-24936238

RESUMEN

Hypoxia inducible factors (HIFs) are transcription factors that activate expression of multiple gene products and promote tumor adaptation to a hypoxic environment. To become transcriptionally active, HIFs associate with cofactors p300 or CBP. Previously, we found that arylsulfonamides can antagonize HIF transcription in a bioassay, block the p300/HIF-1α interaction, and exert potent anticancer activity in several animal models. In the present work, KCN1-bead affinity pull down, (14)C-labeled KCN1 binding, and KCN1-surface plasmon resonance measurements provide initial support for a mechanism in which KCN1 can bind to the CH1 domain of p300 and likely prevent the p300/HIF-1α assembly. Using a previously reported NMR structure of the p300/HIF-1α complex, we have identified potential binding sites in the p300-CH1 domain. A two-site binding model coupled with IC50 values has allowed establishment of a modest ROC-based enrichment and creation of a guide for future analogue synthesis.

20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA