Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Clin Cancer Res ; 30(10): 2272-2285, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38488813

RESUMEN

PURPOSE: Develop and deploy a robust discovery platform that encompasses heterogeneity, clinical annotation, and molecular characterization and overcomes the limited availability of prostate cancer models. This initiative builds on the rich MD Anderson (MDA) prostate cancer (PCa) patient-derived xenograft (PDX) resource to complement existing publicly available databases by addressing gaps in clinically annotated models reflecting the heterogeneity of potentially lethal and lethal prostate cancer. EXPERIMENTAL DESIGN: We performed whole-genome, targeted, and RNA sequencing in representative samples of the same tumor from 44 PDXs derived from 38 patients linked to donor tumor metadata and corresponding organoids. The cohort includes models derived from different morphologic groups, disease states, and involved organ sites (including circulating tumor cells), as well as paired samples representing heterogeneity or stages before and after therapy. RESULTS: The cohort recapitulates clinically reported alterations in prostate cancer genes, providing a data resource for clinical and molecular interrogation of suitable experimental models. Paired samples displayed conserved molecular alteration profiles, suggesting the relevance of other regulatory mechanisms (e.g., epigenomic) influenced by the microenvironment and/or treatment. Transcriptomically, models were grouped on the basis of morphologic classification. DNA damage response-associated mechanisms emerged as differentially regulated between adenocarcinoma and neuroendocrine prostate cancer in a cross-interrogation of PDX/patient datasets. CONCLUSIONS: We addressed the gap in clinically relevant prostate cancer models through comprehensive molecular characterization of MDA PCa PDXs, providing a discovery platform that integrates with patient data and benchmarked to therapeutically relevant consensus clinical groupings. This unique resource supports robust hypothesis generation and testing from basic, translational, and clinical perspectives.


Asunto(s)
Neoplasias de la Próstata , Humanos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Masculino , Animales , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto , Biomarcadores de Tumor/genética , Xenoinjertos , Regulación Neoplásica de la Expresión Génica , Perfilación de la Expresión Génica
2.
Clin Cancer Res ; 26(18): 4933-4946, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32576626

RESUMEN

PURPOSE: Advances in prostate cancer lag behind other tumor types partly due to the paucity of models reflecting key milestones in prostate cancer progression. Therefore, we develop clinically relevant prostate cancer models. EXPERIMENTAL DESIGN: Since 1996, we have generated clinically annotated patient-derived xenografts (PDXs; the MDA PCa PDX series) linked to specific phenotypes reflecting all aspects of clinical prostate cancer. RESULTS: We studied two cell line-derived xenografts and the first 80 PDXs derived from 47 human prostate cancer donors. Of these, 47 PDXs derived from 22 donors are working models and can be expanded either as cell lines (MDA PCa 2a and 2b) or PDXs. The histopathologic, genomic, and molecular characteristics (androgen receptor, ERG, and PTEN loss) maintain fidelity with the human tumor and correlate with published findings. PDX growth response to mouse castration and targeted therapy illustrate their clinical utility. Comparative genomic hybridization and sequencing show significant differences in oncogenic pathways in pairs of PDXs derived from different areas of the same tumor. We also identified a recurrent focal deletion in an area that includes the speckle-type POZ protein-like (SPOPL) gene in PDXs derived from seven human donors of 28 studied (25%). SPOPL is a SPOP paralog, and SPOP mutations define a molecular subclass of prostate cancer. SPOPL deletions are found in 7% of The Cancer Genome Atlas prostate cancers, which suggests that our cohort is a reliable platform for targeted drug development. CONCLUSIONS: The MDA PCa PDX series is a dynamic resource that captures the molecular landscape of prostate cancers progressing under novel treatments and enables optimization of prostate cancer-specific, marker-driven therapy.


Asunto(s)
Antineoplásicos/farmacología , Biomarcadores de Tumor/genética , Medicina de Precisión/métodos , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Humanos , Masculino , Ratones , Cultivo Primario de Células , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Eliminación de Secuencia , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
3.
Endocr Relat Cancer ; 27(7): R255-R265, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32369771

RESUMEN

Many solid tumors metastasize to bone, but only prostate cancer has bone as a single, dominant metastatic site. Recently, the FGF axis has been implicated in cancer progression in some tumors and mounting evidence indicate that it mediates prostate cancer bone metastases. The FGF axis has an important role in bone biology and mediates cell-to-cell communication. Therefore, we discuss here basic concepts of bone biology, FGF signaling axis, and FGF axis function in adult bone, to integrate these concepts in our current understanding of the role of FGF axis in bone metastases.


Asunto(s)
Neoplasias Óseas/secundario , Factores de Crecimiento de Fibroblastos/metabolismo , Femenino , Humanos , Masculino , Transducción de Señal
4.
Clin Cancer Res ; 23(17): 5135-5148, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28512172

RESUMEN

Purpose: Conditioning strategies constitute a relatively unexplored and exciting opportunity to shape tumor fate by targeting the tumor microenvironment. In this study, we assessed how hemin, a pharmacologic inducer of heme oxygenase-1 (HO-1), has an impact on prostate cancer development in an in vivo conditioning model.Experimental Design: The stroma of C57BL/6 mice was conditioned by subcutaneous administration of hemin prior to TRAMP-C1 tumor challenge. Complementary in vitro and in vivo assays were performed to evaluate hemin effect on both angiogenesis and the immune response. To gain clinical insight, we used prostate cancer patient-derived samples in our studies to assess the expression of HO-1 and other relevant genes.Results: Conditioning resulted in increased tumor latency and decreased initial growth rate. Histologic analysis of tumors grown in conditioned mice revealed impaired vascularization. Hemin-treated human umbilical vein endothelial cells (HUVEC) exhibited decreased tubulogenesis in vitro only in the presence of TRAMP-C1-conditioned media. Subcutaneous hemin conditioning hindered tumor-associated neovascularization in an in vivo Matrigel plug assay. In addition, hemin boosted CD8+ T-cell proliferation and degranulation in vitro and antigen-specific cytotoxicity in vivo A significant systemic increase in CD8+ T-cell frequency was observed in preconditioned tumor-bearing mice. Tumors from hemin-conditioned mice showed reduced expression of galectin-1 (Gal-1), key modulator of tumor angiogenesis and immunity, evidencing persistent remodeling of the microenvironment. We also found a subset of prostate cancer patient-derived xenografts and prostate cancer patient samples with mild HO-1 and low Gal-1 expression levels.Conclusions: These results highlight a novel function of a human-used drug as a means of boosting the antitumor response. Clin Cancer Res; 23(17); 5135-48. ©2017 AACR.


Asunto(s)
Galectina 1/genética , Hemo-Oxigenasa 1/genética , Hemina/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/patología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Galectina 1/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/antagonistas & inhibidores , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Masculino , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cell Death Dis ; 7(12): e2570, 2016 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-28032857

RESUMEN

Prostate cancer (PCa) cells display abnormal expression of cytoskeletal proteins resulting in an augmented capacity to resist chemotherapy and colonize distant organs. We have previously shown that heme oxygenase 1 (HO-1) is implicated in cell morphology regulation in PCa. Here, through a multi 'omics' approach we define the HO-1 interactome in PCa, identifying HO-1 molecular partners associated with the integrity of the cellular cytoskeleton. The bioinformatics screening for these cytoskeletal-related partners reveal that they are highly misregulated in prostate adenocarcinoma compared with normal prostate tissue. Under HO-1 induction, PCa cells present reduced frequency in migration events, trajectory and cell velocity and, a significant higher proportion of filopodia-like protrusions favoring zippering among neighboring cells. Moreover forced expression of HO-1 was also capable of altering cell protrusions in transwell co-culture systems of PCa cells with MC3T3 cells (pre-osteoblastic cell line). Accordingly, these effects were reversed under siHO. Transcriptomics profiling evidenced significant modulation of key markers related to cell adhesion and cell-cell communication under HO-1 induction. The integration from our omics-based research provides a four molecular pathway foundation (ANXA2/HMGA1/POU3F1; NFRSF13/GSN; TMOD3/RAI14/VWF; and PLAT/PLAU) behind HO-1 regulation of tumor cytoskeletal cell compartments. The complementary proteomics and transcriptomics approaches presented here promise to move us closer to unravel the molecular framework underpinning HO-1 involvement in the modulation of cytoskeleton pathways, pushing toward a less aggressive phenotype in PCa.


Asunto(s)
Comunicación Celular/genética , Redes Reguladoras de Genes , Hemo-Oxigenasa 1/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Seudópodos/metabolismo , Animales , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Cristalografía por Rayos X , Medios de Cultivo Condicionados/farmacología , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias de la Próstata/genética , Unión Proteica/efectos de los fármacos , Proteómica , Seudópodos/efectos de los fármacos , Análisis de Secuencia de ARN , Espectrometría de Masas en Tándem , Transcriptoma/efectos de los fármacos , Transcriptoma/genética
6.
Oncotarget ; 7(14): 18798-811, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-26933806

RESUMEN

Metabolic syndrome (MeS) has been identified as a risk factor for breast cancer. C-terminal binding protein 1 (CtBP1) is a co-repressor of tumor suppressor genes that is activated by low NAD+/NADH ratio. High fat diet (HFD) increases intracellular NADH. We investigated the effect of CtBP1 hyperactivation by HFD intake on mouse breast carcinogenesis. We generated a MeS-like disease in female mice by chronically feeding animals with HFD. MeS increased postnatal mammary gland development and generated prominent duct patterns with markedly increased CtBP1 and Cyclin D1 expression. CtBP1 induced breast cancer cells proliferation. Serum from animals with MeS enriched the stem-like/progenitor cell population from breast cancer cells. CtBP1 increased breast tumor growth in MeS mice modulating multiple genes and miRNA expression implicated in cell proliferation, progenitor cells phenotype, epithelial to mesenchymal transition, mammary development and cell communication in the xenografts. These results define a novel function for CtBP1 in breast carcinogenesis.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/metabolismo , Síndrome Metabólico/metabolismo , MicroARNs/metabolismo , Animales , Neoplasias de la Mama/genética , Dieta Alta en Grasa , Femenino , Xenoinjertos , Humanos , Células MCF-7 , Síndrome Metabólico/genética , Ratones , Ratones Desnudos , Células 3T3 NIH , Distribución Aleatoria , Factores de Riesgo
7.
Sci Transl Med ; 6(252): 252ra122, 2014 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-25186177

RESUMEN

Bone is the most common site of prostate cancer (PCa) progression to a therapy-resistant, lethal phenotype. We found that blockade of fibroblast growth factor receptors (FGFRs) with the receptor tyrosine kinase inhibitor dovitinib has clinical activity in a subset of men with castration-resistant PCa and bone metastases. Our integrated analyses suggest that FGF signaling mediates a positive feedback loop between PCa cells and bone cells and that blockade of FGFR1 in osteoblasts partially mediates the antitumor activity of dovitinib by improving bone quality and by blocking PCa cell-bone cell interaction. These findings account for clinical observations such as reductions in lesion size and intensity on bone scans, lymph node size, and tumor-specific symptoms without proportional declines in serum prostate-specific antigen concentration. Our findings suggest that targeting FGFR has therapeutic activity in advanced PCa and provide direction for the development of therapies with FGFR inhibitors.


Asunto(s)
Antineoplásicos/uso terapéutico , Bencimidazoles/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Neoplasias de la Próstata/patología , Quinolonas/uso terapéutico , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Bencimidazoles/farmacología , Neoplasias Óseas/patología , Huesos/efectos de los fármacos , Huesos/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Quinolonas/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Clin Cancer Res ; 20(15): 4086-95, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24842953

RESUMEN

PURPOSE: Clinical and epidemiologic data suggest that obesity is associated with more aggressive forms of prostate cancer, poor prognosis, and increased mortality. C-terminal-binding protein 1 (CtBP1) is a transcription repressor of tumor suppressor genes and is activated by NADH binding. High calorie intake decreases intracellular NAD(+)/NADH ratio. The aim of this work was to assess the effect of high-fat diet (HFD) and CtBP1 expression modulation over prostate xenograft growth. EXPERIMENTAL DESIGN: We developed a metabolic syndrome-like disease in vivo model by feeding male nude mice with HFD during 16 weeks. Control diet (CD)-fed animals were maintained at the same conditions. Mice were inoculated with PC3 cells stable transfected with shCtBP1 or control plasmids. Genome-wide expression profiles and Gene Set Enrichment Analysis (GSEA) were performed from PC3.shCtBP1 versus PC3.pGIPZ HFD-fed mice tumors. RESULTS: No significant differences were observed in tumor growth on CD-fed mice; however, we found that only 60% of HFD-fed mice inoculated with CtBP1-depleted cells developed a tumor. Moreover these tumors were significantly smaller than those generated by PC3.pGIPZ control xenografts. We found 823 genes differentially expressed in shCtBP1 tumors from HFD-fed mice. GSEA from expression dataset showed that most of these genes correspond to cell adhesion, metabolic process, and cell cycle. CONCLUSIONS: Metabolic syndrome-like diseases and CtBP1 expression cooperate to induce prostate tumor growth. Hence, targeting of CtBP1 expression might be considered for prostate cancer management and therapy in the subset of patients with metabolic syndromes.


Asunto(s)
Oxidorreductasas de Alcohol/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Dieta Alta en Grasa/efectos adversos , Síndrome Metabólico/prevención & control , Obesidad/prevención & control , Neoplasias de la Próstata/prevención & control , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Adhesión Celular , Proliferación Celular , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Perfilación de la Expresión Génica , Hormonas Esteroides Gonadales/farmacología , Humanos , Técnicas para Inmunoenzimas , Masculino , Síndrome Metabólico/etiología , Síndrome Metabólico/patología , Ratones , Ratones Desnudos , Obesidad/etiología , Obesidad/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Clin Cancer Res ; 18(3): 726-36, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22298898

RESUMEN

PURPOSE: To study Wnt/ß-catenin in castrate-resistant prostate cancer (CRPC) and understand its function independently of the ß-catenin-androgen receptor (AR) interaction. EXPERIMENTAL DESIGN: We carried out ß-catenin immunocytochemical analysis, evaluated TOP-flash reporter activity (a reporter of ß-catenin-mediated transcription), and sequenced the ß-catenin gene in MDA prostate cancer 118a, MDA prostate cancer 118b, MDA prostate cancer 2b, and PC-3 prostate cancer cells. We knocked down ß-catenin in AR-negative MDA prostate cancer 118b cells and carried out comparative gene-array analysis. We also immunohistochemically analyzed ß-catenin and AR in 27 bone metastases of human CRPCs. RESULTS: ß-Catenin nuclear accumulation and TOP-flash reporter activity were high in MDA prostate cancer 118b but not in MDA prostate cancer 2b or PC-3 cells. MDA prostate cancer 118a and MDA prostate cancer 118b cells carry a mutated ß-catenin at codon 32 (D32G). Ten genes were expressed differently (false discovery rate, 0.05) in MDA prostate cancer 118b cells with downregulated ß-catenin. One such gene, hyaluronan synthase 2 (HAS2), synthesizes hyaluronan, a core component of the extracellular matrix. We confirmed HAS2 upregulation in PC-3 cells transfected with D32G-mutant ß-catenin. Finally, we found nuclear localization of ß-catenin in 10 of 27 human tissue specimens; this localization was inversely associated with AR expression (P = 0.056, Fisher's exact test), suggesting that reduced AR expression enables Wnt/ß-catenin signaling. CONCLUSION: We identified a previously unknown downstream target of ß-catenin, HAS2, in prostate cancer, and found that high ß-catenin nuclear localization and low or no AR expression may define a subpopulation of men with bone metastatic prostate cancer. These findings may guide physicians in managing these patients.


Asunto(s)
Glucuronosiltransferasa/genética , Neoplasias de la Próstata/genética , Transducción de Señal/fisiología , beta Catenina/genética , Animales , Western Blotting , Neoplasias Óseas/secundario , Perfilación de la Expresión Génica , Glucuronosiltransferasa/metabolismo , Humanos , Hialuronano Sintasas , Inmunohistoquímica , Masculino , Ratones , Ratones SCID , Mutación , Neoplasias de la Próstata/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo , beta Catenina/metabolismo
10.
Mol Cancer Res ; 9(8): 1078-90, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21700680

RESUMEN

BRCA1 plays numerous roles in the regulation of genome integrity and chemoresistance. Although BRCA1 interaction with key proteins involved in DNA repair is well known, its role as a coregulator in the transcriptional response to DNA damage remains poorly understood. In this study, we show that BRCA1 plays a central role in the transcriptional response to genotoxic stress in prostate cancer. BRCA1 expression mediates apoptosis, cell-cycle arrest, and decreased viability in response to doxorubicin treatment. Xenograft studies using human prostate carcinoma PC3 cells show that BRCA1 depletion results in increased tumor growth. A focused survey of BRCA1-regulated genes in prostate carcinoma reveals that multiple regulators of genome stability and cell-cycle control, including BLM, FEN1, DDB2, H3F3B, BRCA2, CCNB2, MAD2L1, and GADD153, are direct transcriptional targets of BRCA1. Furthermore, we show that BRCA1 targets GADD153 promoter to increase its transcription in response to DNA damage. Finally, GADD153 depletion significantly abrogates BRCA1 influence on cell-cycle progression and cell death in response to doxorubicin treatment. These findings define a novel transcriptional pathway through which BRCA1 orchestrates cell fate decisions in response to genotoxic insults, and suggest that BRCA1 status should be considered for new chemotherapeutic treatment strategies in prostate cancer.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Proteína BRCA1/metabolismo , Carcinoma/metabolismo , Daño del ADN/genética , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/genética , Neoplasias de la Próstata/metabolismo , Factor de Transcripción CHOP/metabolismo , Animales , Proteína BRCA1/genética , Carcinoma/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Inestabilidad Genómica/genética , Humanos , Masculino , Ratones , Neoplasias de la Próstata/patología , Factor de Transcripción CHOP/genética
11.
Mol Cancer Res ; 7(11): 1745-55, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19903769

RESUMEN

Prostate cancer (PCa) is the second leading cause of cancer-associated death in men. Inflammation has been recognized as a risk factor for this disease. Heme oxygenase 1 (HO-1), the inducible isoform of the rate-limiting enzyme in heme degradation, counteracts oxidative and inflammatory damage. Here, we investigated the regulated expression of HO-1 and its functional consequences in PCa. We studied the effect of genetic and pharmacologic disruption of HO-1 in the growth, invasion, and migration in androgen-sensitive (MDA PCa2b and LNCaP) and androgen-insensitive (PC3) PCa cell lines. Our results show that HO-1 levels are markedly decreased in PC3 compared with MDA PCa2b and LNCaP. Hemin treatment increased HO-1 at both protein and mRNA levels in all cell lines and decreased cell proliferation and invasion. Furthermore, overexpression of HO-1 in PC3 resulted in markedly reduced cell proliferation and migration. Accordingly, small interfering RNA-mediated silencing of HO-1 expression in MDA PCa2b cells resulted in increased proliferation and invasion. Using reverse transcription-quantitative PCR-generated gene array, a set of inflammatory and angiogenic genes were upregulated or downregulated in response to HO-1 overexpression identifying matrix metalloprotease 9 (MMP9) as a novel downstream target of HO-1. MMP9 production and activity was downregulated by HO-1 overexpression. Furthermore, PC3 cells stably transfected with HO-1 (PC3HO-1) and controls were injected into nu/nu mice for analysis of in vivo tumor xenograft phenotype. Tumor growth and MMP9 expression was significantly reduced in PC3HO-1 tumors compared with control xenografts. Taken together, these results implicate HO-1 in PCa cell migration and proliferation suggesting its potential role as a therapeutic target in clinical settings.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Animales , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Hemo-Oxigenasa 1/genética , Hemina/farmacología , Humanos , Inmunohistoquímica , Masculino , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Análisis por Micromatrices , Invasividad Neoplásica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transfección , Trasplante Heterólogo
12.
J Clin Invest ; 118(8): 2697-710, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18618013

RESUMEN

In prostate cancer, androgen blockade strategies are commonly used to treat osteoblastic bone metastases. However, responses to these therapies are typically brief, and the mechanism underlying androgen-independent progression is not clear. Here, we established what we believe to be the first human androgen receptor-negative prostate cancer xenografts whose cells induced an osteoblastic reaction in bone and in the subcutis of immunodeficient mice. Accordingly, these cells grew in castrated as well as intact male mice. We identified FGF9 as being overexpressed in the xenografts relative to other bone-derived prostate cancer cells and discovered that FGF9 induced osteoblast proliferation and new bone formation in a bone organ assay. Mice treated with FGF9-neutralizing antibody developed smaller bone tumors and reduced bone formation. Finally, we found positive FGF9 immunostaining in prostate cancer cells in 24 of 56 primary tumors derived from human organ-confined prostate cancer and in 25 of 25 bone metastasis cases studied. Collectively, these results suggest that FGF9 contributes to prostate cancer-induced new bone formation and may participate in the osteoblastic progression of prostate cancer in bone. Androgen receptor-null cells may contribute to the castration-resistant osteoblastic progression of prostate cancer cells in bone and provide a preclinical model for studying therapies that target these cells.


Asunto(s)
Neoplasias Óseas/secundario , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Osteogénesis/genética , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Cariotipificación , Queratinas/metabolismo , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Metástasis de la Neoplasia , Trasplante de Neoplasias , Orquiectomía , Técnicas de Cultivo de Órganos , Osteoblastos/metabolismo , Neoplasias de la Próstata/genética , Trasplante Heterólogo , Vimentina/metabolismo
13.
BMC Cancer ; 6: 286, 2006 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-17169158

RESUMEN

BACKGROUND: Chronic injury deregulates cellular homeostasis and induces a number of alterations leading to disruption of cellular processes such as cell cycle checkpoints and apoptosis, driving to carcinogenesis. The stress protein heme oxygenase-1 (HO-1) catalyzes heme degradation producing biliverdin, iron and CO. Induction of HO-1 has been suggested to be essential for a controlled cell growth. The aim of this work was to analyze the in vivo homeostatic response (HR) triggered by the withdrawal of a potent carcinogen, p-dimethylaminoazobenzene (DAB), after preneoplastic lesions were observed. We analyzed HO-1 cellular localization and the expression of HO-1, Bcl-2 and cell cycle related proteins under these conditions comparing them to hepatocellular carcinoma (HC). METHODS: The intoxication protocol was designed based on previous studies demonstrating that preneoplastic lesions were evident after 89 days of chemical carcinogen administration. Male CF1 mice (n = 18) were used. HR group received DAB (0.5 % w/w) in the diet for 78 days followed by 11 days of carcinogen deprivation. The HC group received the carcinogen and control animals the standard diet during 89 days. The expression of cell cycle related proteins, of Bcl-2 and of HO-1 were analyzed by western blot. The cellular localization and expression of HO-1 were detected by immnunohistochemistry. RESULTS: Increased expression of cyclin E/CDK2 was observed in HR, thus implicating cyclin E/CDK2 in the liver regenerative process. p21cip1/waf1 and Bcl-2 induction in HC was restituted to basal levels in HR. A similar response profile was found for HO-1 expression levels, showing a lower oxidative status in the carcinogen-deprived liver. The immunohistochemical studies revealed the presence of macrophages surrounding foci of necrosis and nodular lesions in HR indicative of an inflammatory response. Furthermore, regenerative cells displayed changes in type, size and intensity of HO-1 immunostaining. CONCLUSION: These results demonstrate that the regenerative capacity of the liver is still observed in the pre-neoplastic tissue after carcinogen withdrawal suggesting that reversible mechanism/s to compensate necrosis and to restitute homeostasis are involved.


Asunto(s)
Carcinógenos/toxicidad , Proteínas de Ciclo Celular/genética , Ciclo Celular/efectos de los fármacos , Hemo-Oxigenasa 1/genética , p-Dimetilaminoazobenceno/toxicidad , Animales , Carcinógenos/administración & dosificación , Carcinoma Hepatocelular/enzimología , Esquema de Medicación , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Homeostasis , Neoplasias Hepáticas/enzimología , Masculino , Ratones , Ratones Endogámicos , p-Dimetilaminoazobenceno/administración & dosificación
14.
Per Med ; 1(1): 127-130, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29793225

RESUMEN

Genomics must be combined with proteomics and metabolomics to rationalize a therapeutic strategy that considers gene expression, protein expression and metabolic profiles in the target organ to gain insight into other pathways implicated in the same or contributory tissues. Multidisciplinary strategies such as this provide an interactive process by which findings are translated into novel therapies.

15.
Carcinogenesis ; 23(8): 1289-96, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12151346

RESUMEN

Current therapy for advanced prostate cancer is largely based on androgen deprivation and is mostly palliative because all patients eventually relapse with androgen-independent disease. Doxorubicin (Dx), an anthracycline used commonly as a chemotherapeutic agent in relapsed prostate cancer, is a strong inducer of p53 expression and p21(CIP1/WAF1) (p21) transactivation. Previous reports suggest that p21 may have a role in the modulation to chemotherapy-induced apoptosis, prostate cancer progression and androgen regulation. In order to investigate if p21 has a pro-survival role in the response of prostate cancer cells to cellular stress, we exposed two androgen-regulated human prostate cancer cell lines (MDA PCa 2b and LNCaP) to Dx and growth factor withdrawal. We then studied expression of p53 and p21, cell-cycle kinetics and apoptosis. We have found that p53 protein accumulated in a dose- and time-dependent manner after Dx treatment, while p21 expression increased over time with low but decreased with high Dx doses. Apoptosis occurred in parallel with p21 down-modulation. Dx treatment of p53 knockout cells demonstrated that p21 induction was strictly p53 dependent. Reduction of p21 levels in prostate cancer cells with an antisense p21 adenovirus resulted in sensitization to Dx and accelerated onset of apoptosis in response to growth factor withdrawal. The evidence presented here also suggests that caspase activation mediates the apoptosis in this system and supports that p21 may modulate the threshold of apoptosis in prostate cancer. These observations may thus provide implications onto the integration of chemotherapy and androgen ablation.


Asunto(s)
Apoptosis/fisiología , Ciclinas/fisiología , Daño del ADN , Sustancias de Crecimiento/metabolismo , Neoplasias Hormono-Dependientes/patología , Neoplasias de la Próstata/patología , Antibióticos Antineoplásicos/farmacología , Ciclo Celular , Supervivencia Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Doxorrubicina/farmacología , Humanos , Masculino , Neoplasias Hormono-Dependientes/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
16.
BMC Cancer ; 2: 6, 2002 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-11914144

RESUMEN

BACKGROUND: Acute Intermittent Porphyria is a genetic disorder of heme metabolism, characterized by increased levels of porphyrin precursors, delta-aminolevulinic acid (ALA) and porphobilinogen (PBG). ALA has been reported to generate reactive oxygen species and to cause oxidative damage to proteins, subcellular structures and DNA. It is known that oxidative stress can induce apoptosis. The aim of this work was to study the cytotoxic effect of ALA on two hepatocarcinoma cell lines. RESULTS: We have determined the impact of ALA on HEP G2 and HEP 3B hepatocarcinoma cell lines survival as measured by the MTT assay. ALA proved to be cytotoxic in both cell lines however; HEP G2 was more sensitive to ALA than HEP 3B. Addition of hemin or glucose diminished ALA cytotoxicity in HEP G2 cells; instead it was enhanced in HEP 3B cells. Because apoptosis is usually associated with DNA fragmentation, the DNA of ALA treated and untreated cells were analyzed. The characteristic pattern of DNA fragmentation ladders was observed in ALA treated cells. To elucidate the mechanisms of ALA induced apoptosis, we examined its effect on p53 expression. No changes in p53 mRNA levels were observed after exposure of both cell lines to ALA for 24 h. CDK2 and CDK4 protein levels were reduced after ALA treatment at physiological concentrations.


Asunto(s)
Ácido Aminolevulínico/toxicidad , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Ácido Aminolevulínico/uso terapéutico , Carcinoma Hepatocelular/metabolismo , Ciclo Celular/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , ADN de Neoplasias/metabolismo , Relación Dosis-Respuesta a Droga , Glucosa/farmacología , Hemina/farmacología , Humanos , Neoplasias Hepáticas/metabolismo , Sales de Tetrazolio/metabolismo , Tiazoles/metabolismo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...