Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
1.
Br J Cancer ; 109(12): 3105-15, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24157827

RESUMEN

BACKGROUND: Glutathione S-transferase mu 3 (GSTM3) has been proven to be downregulated in renal cell carcinoma (RCC). We aimed to characterise the role of GSTM3 and its genetic predisposition on the occurrence and postoperative prognosis of RCC. METHODS: The effect of GSTM3 on RCC aggressiveness was examined using transfection and silencing methods. Glutathione S-transferase mu 3 expression in renal tissues was examined by immunohistochemistry. The associations of rs1332018 (A-63C) and rs7483 (V224I) polymorphisms with RCC risk were examined using 400 RCC patients and 802 healthy controls. The factors contributing to postoperative disease-specific survival of RCC patients were evaluated using the Cox proportional hazard model. RESULTS: Glutathione S-transferase mu 3 silencing increased the invasion and anchorage-independent growth of RCC cell lines. rs1332018 (AC+CC vs AA), which correlated with low expression of GSTM3 in kidney, was associated with RCC risk (odds ratio, 1.446; 95% confidence interval (CI), 1.111-1.882). rs1332018 variants and low GSTM3 expression significantly predicted unfavourable postoperative survivals of RCC patients (P<0.05). rs1332018 variants independently predicted a poor prognosis (hazard ratio, 2.119; 95% CI, 1.043-4.307). CONCLUSION: Glutathione S-transferase mu 3 may function as a tumour suppressor in RCC. rs1332018 genetic variants predispose the host to downregulating GSTM3 expression in kidney, facilitate carcinogenesis, and predict an unfavourable postoperative prognosis of RCC.


Asunto(s)
Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/genética , Glutatión Transferasa/genética , Neoplasias Renales/enzimología , Neoplasias Renales/genética , Carcinoma de Células Renales/patología , Estudios de Casos y Controles , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Predisposición Genética a la Enfermedad , Variación Genética , Glutatión Transferasa/biosíntesis , Glutatión Transferasa/deficiencia , Glutatión Transferasa/metabolismo , Humanos , Inmunohistoquímica , Neoplasias Renales/patología , Masculino , Polimorfismo de Nucleótido Simple , Pronóstico , Regiones Promotoras Genéticas , Factores de Riesgo , Transfección
2.
Gene Ther ; 18(10): 969-78, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21512508

RESUMEN

We previously identified the mouse and human Glipr1 and GLIPR1/RTVP-1 genes, respectively, as direct p53 targets with proapoptotic activities in various cancer cell lines, including prostate cancer (PCa). Intratumoral injection of an adenoviral vector capable of efficient transduction and expression of Glipr1 (AdGlipr1) yielded promising therapeutic results in an orthotopic, metastatic mouse model of PCa. AdGlipr1-transduced macrophages (Mφ/Glipr1) generated greater surface expression of CD40, CD80 and major histocompatibility complex class II molecules and greater production of interleukin 12 (IL-12) and IL-6 in vitro than control macrophages did. Mechanistic analysis indicated that increased production of IL-12 in Mφ/Glipr1 depends on activation of the p38 signaling cascade. Mφ/Glipr1 injected into orthotopic 178-2BMA tumors in vivo resulted in significantly suppressed prostate tumor growth and spontaneous lung metastases and longer survival relative to those observed in control-treated mice. Furthermore, these preclinical data indicate the generation of systemic natural killer cell activity and tumor-specific cytotoxic T lymphocyte responses. Trafficking studies confirmed that intratumorally injected Mφ/Glipr1 could migrate to draining lymph nodes. Overall, our data suggest that this novel gene-modified cell approach is an effective treatment avenue that induces antitumor immune responses in preclinical studies.


Asunto(s)
Terapia Genética/métodos , Macrófagos/metabolismo , Metástasis de la Neoplasia/prevención & control , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia , Proteínas/genética , Adenoviridae , Animales , Antígeno B7-1/metabolismo , Antígenos CD40/metabolismo , Vectores Genéticos/administración & dosificación , Interleucina-12/metabolismo , Interleucina-6/metabolismo , Células Asesinas Naturales/inmunología , Cinética , Masculino , Ratones , Neoplasias de la Próstata/inmunología , Linfocitos T Citotóxicos/inmunología
3.
Nat Med ; 4(9): 1062-4, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9734401

RESUMEN

Although prostate cancer cells are often initially sensitive to androgen ablation, they eventually lose this response and continue to survive, grow and spread in the absence of androgenic steroids. The mechanism(s) that underlie resistance to androgen ablation therapy remain mostly unknown. We have demonstrated that elevated caveolin protein levels are associated with human prostate cancer progression in pathological specimens. Here we show that suppression of caveolin expression by a stably transfected antisense caveolin-1 cDNA vector converted androgen-insensitive metastatic mouse prostate cancer cells to an androgen-sensitive phenotype. Orthotopically grown tumors and low-density cell cultures derived from antisense caveolin clones had increased apoptosis in the absence of androgenic steroids, whereas similarly grown tumors and cells from vector (control) clones and parental cells were not sensitive to androgens. Studies using a representative antisense caveolin clone showed that selection for androgen resistance in vivo correlated with increased caveolin levels, and that adenovirus-mediated caveolin expression blocked androgen sensitivity. Our results identify a new candidate gene for hormone-resistant prostate cancer in man and indicate that androgen insensitivity can be an inherent property of metastatic prostate cancer.


Asunto(s)
Andrógenos/metabolismo , Caveolinas , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Apoptosis , Caveolina 1 , Modelos Animales de Enfermedad , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Conejos , Testosterona/farmacología , Células Tumorales Cultivadas
4.
J Cell Biol ; 96(6): 1671-6, 1983 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-6853598

RESUMEN

Adult bladder epithelium (BLE) is induced to differentiate into glandular epithelium after association with urogenital sinus mesenchyme (UGM) and subsequent in vivo growth in syngeneic male hosts. Alteration of epithelial cytodifferentiation is associated with the expression of prostate-specific antigens, histochemical and steroid metabolic activities. These observations suggest that the inductive influence of the UGM has reprogrammed both the morphological and functional characteristics of the urothelium. In this report, differences regarding the mechanisms and effects of androgenic stimulation of prostate and bladder are exploited to determine the extent to which UGM plus BLE recombinants express a prostatelike, androgen-dependent phenotype. Results from cytosolic and autoradiographic binding studies suggest that androgen binding is induced in UGM plus BLE recombinants and that this activity is accounted for by the induced urothelial cells. In UGM plus BLE recombinants, androgen-induced [3H]thymidine or [35S]-methionine uptake analyzed by two-dimensional gel electrophoresis was qualitatively and quantitatively similar to that of prostate as opposed to bladder. These studies indicate that expression within BLE of prostatic phenotype is associated with a loss of urothelial characteristics and that androgen sensitivity is presumably a function of the inductive activities of the stroma.


Asunto(s)
Próstata/citología , Vejiga Urinaria/citología , Animales , Diferenciación Celular , División Celular , Replicación del ADN , Electroforesis en Gel de Poliacrilamida , Células Epiteliales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Embarazo , Ratas , Receptores Androgénicos/análisis
5.
Gene Ther ; 15(10): 787-96, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18385769

RESUMEN

There is a critical need to develop new and effective cancer therapies that target bone, the primary metastatic site for prostate cancer and other malignancies. Among the various therapeutic approaches being considered for this application, gene-modified cell-based therapies may have specific advantages. Gene-modified cell therapy uses gene transfer and cell-based technologies in a complementary fashion to chaperone appropriate gene expression cassettes to active sites of tumor growth. In this paper, we briefly review potential cell vehicles for this approach and discuss relevant gene therapy strategies for prostate cancer. We further discuss selected studies that led to the conceptual development and preclinical testing of IL-12 gene-modified bone marrow cell therapy for prostate cancer. Finally, we discuss future directions in the development of gene-modified cell therapy for metastatic prostate cancer, including the need to identify and test novel therapeutic genes such as GLIPR1.


Asunto(s)
Trasplante de Médula Ósea/métodos , Neoplasias Óseas/secundario , Neoplasias Óseas/terapia , Terapia Genética/métodos , Neoplasias de la Próstata/terapia , Células de la Médula Ósea/inmunología , Ingeniería Genética , Humanos , Inmunoterapia/métodos , Interleucina-12/genética , Masculino , Viroterapia Oncolítica/métodos
6.
Cancer Gene Ther ; 14(10): 819-27, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17627292

RESUMEN

To investigate the immunomodulatory effects of interleukin-12 (IL-12) for treatment of metastatic prostate cancer, we administered adult bone marrow cells (BMC) that were genetically modified by retroviral vector-mediated IL-12 gene transduction in an experimental mouse model of prostate cancer metastasis. This therapy produced significant anti-metastatic effects in bone and lung and prolonged animal survival. Flow cytometric analysis indicated donor BMC could effectively home to bone and lung after treatment. Intensive infiltration of CD4 and CD8T cells in lung metastases and increased systemic natural killer and cytotoxic T lymphocyte activities indicated induction of a significant anti-metastatic immune response after treatment with IL-12 transduced BMC. Our results demonstrate the therapeutic potential of gene-modified BMC gene therapy.


Asunto(s)
Células de la Médula Ósea/fisiología , Neoplasias Óseas/prevención & control , Modelos Animales de Enfermedad , Interleucina-12/genética , Neoplasias Pulmonares/prevención & control , Neoplasias de la Próstata/prevención & control , Animales , Neoplasias Óseas/inmunología , Neoplasias Óseas/secundario , Expresión Génica , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Masculino , Ratones , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Retroviridae/genética , Tasa de Supervivencia , Linfocitos T Citotóxicos/inmunología , Transducción Genética
7.
Mol Biol Cell ; 10(2): 407-16, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9950685

RESUMEN

The type IV collagenases/gelatinases matrix metalloproteinase-2 (MMP-2) and MMP-9 play a variety of important roles in both physiological and pathological processes and are regulated by various growth factors, including transforming growth factor-beta1 (TGF-beta1), in several cell types. Previous studies have suggested that cellular control of one or both collagenases can occur through direct transcriptional mechanisms and/or after secretion through proenzyme processing and interactions with metalloproteinase inhibitors. Using human prostate cancer cell lines, we have found that TGF-beta1 induces the MMP-9 proenzyme; however, this induction does not result from direct effects on gene transcription but, instead, through a protein synthesis-requiring process leading to increased MMP-9 mRNA stability. In addition, we have examined levels of TGF-beta1 regulation of MMP-2 in one prostate cancer cell line and found that TGF-beta1 induces higher secreted levels of this collagenase through increased stability of the secreted 72-kDa proenzyme. These results identify two novel nontranscriptional pathways for the cellular regulation of MMP-9 and MMP-2 collagenase gene expression and activities.


Asunto(s)
Colagenasas/metabolismo , Gelatinasas/metabolismo , Metaloendopeptidasas/metabolismo , Neoplasias de la Próstata/enzimología , Factor de Crecimiento Transformador beta/farmacología , Colagenasas/genética , Cicloheximida/farmacología , Dactinomicina/farmacología , Gelatinasas/genética , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Metaloendopeptidasas/genética , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Neoplasias de la Próstata/genética , Inhibidores de la Síntesis de la Proteína/farmacología , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Activación Transcripcional/efectos de los fármacos , Células Tumorales Cultivadas
8.
Cancer Gene Ther ; 13(1): 91-8, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16052232

RESUMEN

We developed a preclinical prostate cancer model to study the feasibility of adoptive immunotherapy for residual tumor following neo-adjuvant in situ adenoviral-vector-mediated interleukin 12 (AdIL-12) gene therapy. Splenocytes were obtained from mice with orthotopic 178-2 BMA metastatic mouse prostate cancers treated previously with AdIL-12, or a vector with the IL-12 genes plus the costimulatory gene B7-1 (AdIL-12/B7), or a control gene (Adbetagal). The splenocytes were subsequently injected intravenously into syngeneic mice bearing orthotopic 178-2 BMA tumors generated 3 days previously. Significant orthotopic tumor growth suppression was achieved with splenocytes derived from mice whose tumors had been injected with AdIL-12 compared to splenocytes from control Adbetagal mice (P = 0.0005) and splenocytes from AdIL-12/B7-treated mice significantly suppressed spontaneous lung metastases compared to splenocytes from control mice (P = 0.0356). Adoptive transfer of splenocytes from either AdIL-12 (P = 0.004) or AdIL-12/B7 (P = 0.009)-treated mice significantly prolonged survival relative to controls. Transfer of NK and tumor-specific CTL activities was detected and depletion of CD4+ and CD8+ T cells by in vitro antibody-mediated complement lysis of the splenocytes prior to injection abrogated the effects. Systemic IL-12 administration delivered by intramuscular AdIL-12 injection enhanced the antitumor effects of adoptive splenocyte transfer and boosted the CTL response. Our data provide evidence that this form of adoptive immunotherapy can enhance the effectiveness of neo-adjuvant in situ IL-12 gene therapy in cases of persistent malignancy.


Asunto(s)
Traslado Adoptivo/métodos , Terapia Genética , Interleucina-12/metabolismo , Neoplasias de la Próstata/terapia , Bazo/metabolismo , Animales , Antígeno B7-1/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Inmunoterapia , Masculino , Ratones , Metástasis de la Neoplasia , Neoplasias de la Próstata/metabolismo , Bazo/citología , Linfocitos T/metabolismo , Factores de Tiempo
9.
Cancer Gene Ther ; 13(7): 658-63, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16485011

RESUMEN

We previously identified a novel p53 target gene, RTVP-1, that possesses unique cytotoxic and immunostimulatory activities which make it potentially useful for cancer gene therapy. To test the therapeutic potential of RTVP-1 in a gene-modified tumor cell-based vaccine model, we used an adenoviral vector capable of efficient transduction and expression of RTVP-1 (AdRTVP-1), together with a highly metastatic mouse prostate cancer cell line (178-2 BMA). A vaccine was prepared with 178-2 BMA cells transduced with AdRTVP-1 or a control adenoviral vector expressing beta-galactosidase (Adbetagal). After irradiation of the cells, syngeneic 129/Sv mice were vaccinated three times at weekly intervals. After 3 weeks, they were challenged with orthotopic 178-2 BMA cells. After 21 days, fewer than 60% of the RTVP-1-cell-vaccinated mice developed tumors compared to 100% of the control mice. The RTVP-1-cell vaccine significantly reduced primary tumor wet weight compared with control Adbetagal-cell vaccine (P<0.0001 at 7 and 14 days). Experimental metastasis to lung was also significantly reduced (P=0.0377), and survival significantly increased (P=0.0002). In addition, significantly increased NK and CTL activities were demonstrated in the AdRTVP-1-cell-vaccinated mice. These findings indicate that RTVP-1 gene-modified cell-based vaccines may be useful in the prevention of recurrent prostate cancer.


Asunto(s)
Adenoviridae , Vacunas contra el Cáncer/uso terapéutico , Proteínas de Neoplasias , Proteínas del Tejido Nervioso , Neoplasias de la Próstata/prevención & control , Animales , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Humanos , Masculino , Proteínas de la Membrana , Ratones , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Proteínas del Tejido Nervioso/inmunología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Transducción Genética
10.
J Natl Cancer Inst ; 93(13): 998-1007, 2001 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-11438565

RESUMEN

BACKGROUND: Immunization with modified tumor cells carrying recombinant immunomodulatory genes is being explored as cancer immunotherapy. In this study, we examine whether canarypox ALVAC viruses carrying immunostimulatory cytokine genes (granulocyte-macrophage colony-stimulating factor, interleukin 2, interleukin 12, and tumor necrosis factor-alpha) can induce antitumor immunity (to rechallenge) in the RM-1 model of a highly aggressive, weakly immunogenic murine prostate cancer. METHODS: For antitumor activity studies, RM-1 murine prostate cancer cells were infected with the parental ALVAC virus or one or two recombinant ALVAC-cytokine viruses and then injected into male C57BL/6 mice. For rechallenge studies, other mice were first given an injection subcutaneously with irradiated (nonproliferating) recombinant ALVAC-infected RM-1 cells and then (10 days later) with untreated RM-1 cells. For the determination of which immune cells were required for antitumor activity, mice were immunodepleted of CD4, CD8, or natural killer (NK) NK1.1 cells with the corresponding monoclonal antibodies and were then given an injection of ALVAC-cytokine-infected RM-1 cells. For all experiments, tumor outgrowth and animal survival were monitored. RESULTS: After subcutaneous injection into mice, RM-1 cells infected with one (except ALVAC-interleukin 2) or two ALVAC-cytokine recombinants had statistically significantly greater antitumor activity than RM-1 cells infected with parental ALVAC (P<.001 for all; two-sided test). The antitumor activity of RM-1 cells infected with any two ALVAC-cytokine recombinants was greater than, but not statistically significantly different from, that of RM-1 cells infected with any one ALVAC-cytokine recombinant. NK1.1 cells were necessary for antitumor activity, but tumor-specific CD4(+) regulatory T cells were also induced that inhibited CD8(+) RM-1-specific cytotoxic T cells, resulting in the lack of immunity to a rechallenge by RM-1 cells. DISCUSSION: Canarypox viruses can transfer immunostimulatory cytokine genes into RM-1 prostate cancer cells. When such cells were injected into mice, the cytokines induced an antitumor response against this highly aggressive, weakly immunogenic tumor. This response, however, did not protect the mouse against a rechallenge with RM-1 cells because suppressor CD4(+) T cells were induced that inhibited tumor-specific CD8(+) cytotoxic T cells.


Asunto(s)
Avipoxvirus/genética , Neoplasias de la Próstata/terapia , Proteínas , Animales , Anticuerpos Monoclonales/metabolismo , Antígenos/biosíntesis , Antígenos Ly , Antígenos de Superficie , Antígenos CD4/biosíntesis , Antígenos CD8/biosíntesis , Citometría de Flujo , Técnicas de Transferencia de Gen , Terapia Genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Interleucina-12/genética , Interleucina-2/genética , Lectinas Tipo C , Masculino , Ratones , Ratones Endogámicos C57BL , Subfamilia B de Receptores Similares a Lectina de Células NK , Trasplante de Neoplasias , Neoplasias de la Próstata/inmunología , Biosíntesis de Proteínas , Proteínas Recombinantes/metabolismo , Linfocitos T Citotóxicos/metabolismo , Factores de Tiempo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/genética
11.
Cancer Res ; 58(19): 4288-91, 1998 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-9766654

RESUMEN

The type IV collagenases matrix metalloprotease (MMP)-2 and MMP-9 are linked with a wide array of biological activities, including tumor invasion, metastasis, and angiogenesis. Here, we report that neuropeptide hormones, which are present in prostatic adenocarcinomas, can stimulate secreted activity of MMP-9 in human prostate cancer cell lines. Northern blotting analyses demonstrated that neuropeptide stimulation lead to elevated mRNA levels of MMP-9 but not MMP-2. Further assays of MMP-9 promoter activation and a nuclear run-off indicated that neuropeptide induction of MMP-9 expression occurs at the level of transcription. These data indicate that neuropeptides can regulate MMP activity, which, in turn, could facilitate prostate cancer progression.


Asunto(s)
Colagenasas/biosíntesis , Neuropéptidos/farmacología , Transcripción Genética/efectos de los fármacos , Adenocarcinoma , Núcleo Celular/metabolismo , Cloranfenicol O-Acetiltransferasa/biosíntesis , Colagenasas/genética , Inducción Enzimática , Gelatinasas/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Metaloendopeptidasas/biosíntesis , Peso Molecular , Regiones Promotoras Genéticas , Neoplasias de la Próstata , ARN Mensajero/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Transfección , Células Tumorales Cultivadas
12.
Cancer Res ; 44(3): 1019-26, 1984 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-6692389

RESUMEN

Because sodium molybdate stabilizes steroid receptors, this compound has been included in the homogenizing medium in order to maximize the recovery of measurable steroid receptors in normal and neoplastic tissues. This study demonstrates that sodium molybdate extracts additional androgen receptors from prostatic nuclei in a concentration-dependent manner. Nuclei previously washed with Triton X-100 to remove the outer nuclear membranes released similar numbers of androgen receptors with sodium molybdate as the unwashed nuclei, suggesting that the extracted nuclear androgen receptors are associated with intranuclear matrices. Sucrose density gradient analyses revealed that sodium molybdate-extractable nuclear androgen receptors sedimented similarly to the 0.4 M KCl extract as 4S receptor complexes under high-salt conditions. We have compared the amount of nuclear androgen receptors extracted from normal prostates (ventral prostate and dorsolateral prostate) and Noble hooded rat and Dunning prostatic tumors by a sensitive translocation-extraction procedure. This procedure involves the incubation of minced prostatic tissues, isolated from castrated rats, with [3H]R1881 ( [3H]methyltrienolone; [6,7-3H]-17 beta-hydroxy-17 alpha-methylestra-4,9,11-trien-3-one) at 37 degrees for 2 hr. Crude nuclear pellets were prepared from the minced tissues, and nuclear androgen receptors were extracted with 40 mM Na2MoO4 or 0.4 M KCl. Results showed that the amount of nuclear androgen receptors present in the prostatic tumor nuclei is lower than that found in the normal. Although the percentage of nuclear androgen receptors extracted by sodium molybdate or KCl is similar between androgen-dependent and androgen-independent prostatic tumors, the absolute amounts of nuclear androgen receptors per mg DNA extracted from the former are 2- to 8-fold higher than those found in the latter.


Asunto(s)
Núcleo Celular/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/aislamiento & purificación , Receptores de Esteroides/aislamiento & purificación , Animales , Castración , Citosol/metabolismo , Femenino , Cinética , Masculino , Molibdeno , Ratas , Ratas Endogámicas , Receptores Androgénicos/metabolismo , Especificidad de la Especie
13.
Cancer Res ; 60(7): 1927-33, 2000 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10766182

RESUMEN

Better understanding of the immunology of prostate cancer is needed for the development of new therapeutic approaches that can be used in conjunction with current treatment methods. The present study was designed to compare the immunological properties of a genetically matched pair of primary tumor- and metastasis-derived prostate cancer cell lines generated from the mouse prostate reconstitution (MPR) model. Only the primary prostate cancer cells were immunogenic in that prior immunization with irradiated primary but not the metastatic prostate cancer cells delayed the growth of subsequently injected live cancer cells. The lack of immunogenicity of the metastatic cells was not attributable to their inability to induce antitumor cytotoxic T cells. Both primary and metastatic cells induced antitumor CTLs in syngeneic hosts, but unlike the primary cells, the metastatic cells were resistant to CTL lysis. Differential resistance to cytolysis in metastatic versus primary prostate cancer cells was not attributable to the differential expression of molecules such as transporter associated with antigen processing (TAP)-1, TAP-2, low molecular weight protein of the proteasome complex (LMP)-2, and LMP-7 that contribute to antigen presentation by class I MHC. IFN-gamma induced surface class I MHC expression, as well as gene expression of TAP-1, TAP-2, LMP-2, and LMP-7 in the metastatic cells, yet the cells remained resistant to cell lysis induced by CTLs. Interestingly, although in comparison to the primary cells the metastatic cells were resistant to cytolysis, both cell types were susceptible to DNA fragmentation induced by CTLs. Cell fusion between primary and metastatic cancer cells resulted in hybrids that also resisted the cytolytic activity of CTLs. Therefore, there is a dominant factor(s) in the metastatic prostate cancer cells that confers specific protection against CTL cytolysis in this model system.


Asunto(s)
Citotoxicidad Inmunológica , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Linfocitos T Citotóxicos/inmunología , Animales , Fusión Celular , Antígenos de Histocompatibilidad Clase I/inmunología , Interferón gamma/farmacología , Interleucina-2/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Proteínas Recombinantes , Linfocitos T Citotóxicos/efectos de los fármacos , Factores de Tiempo , Células Tumorales Cultivadas
14.
Cancer Res ; 56(14): 3359-65, 1996 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-8764134

RESUMEN

Tumor progression to the stage of metastasis may result in part from the selection of certain primary tumor cell clones which are phenotypically competent for survival, invasion, and growth at secondary sites. Selection for traits such as loss of growth inhibitory responses, acquisition of increased adhesiveness, increased local immunosuppression, and enhanced motility and collagenase activities likely contribute to cancer progression and may be regulated through the action of growth factors. The transforming growth factors (TGF-beta) family of growth factors has often been associated with these traits and tumor progression; therefore, elimination or subversion of TGF-beta-responsive pathways should be considered as a mechanistic framework for metastatic events. In this report, we have compared growth and extracellular matrix responses to TGF-beta in six metastatic and six primary tumor-derived cell lines in a mouse model of prostate cancer. We have found that tumor cell lines derived from focal pulmonary metastasis secreted relatively greater quantities of total TGF-betas, lost most or all TGF-beta1 growth inhibition, but responded to TGF-beta1 through induction of the type IV collagenase matrix metalloproteinase-9, whereas cell lines derived from tumors which proliferated at the primary site retained the growth inhibition but lacked collagenase activity. Synthesis of another extracellular matrix protein, plasminogen activator inhibitor 1, was stimulated by TGF-beta1 in both primary as well as metastatic tumors. These results suggest that acquisition of differential responses to the TGF-beta family could result in phenotypic traits which facilitate tumor metastasis from certain primary site clones.


Asunto(s)
Metástasis de la Neoplasia , Neoplasias de la Próstata/patología , Factor de Crecimiento Transformador beta/farmacología , Animales , División Celular/efectos de los fármacos , Colagenasas/metabolismo , Inducción Enzimática , Gelatinasas/metabolismo , Genes myc , Genes p53 , Genes ras , Neoplasias Pulmonares/secundario , Masculino , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Metaloendopeptidasas/metabolismo , Ratones , Ratones Noqueados , Peso Molecular , Inhibidor 1 de Activador Plasminogénico/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Células Tumorales Cultivadas
15.
Cancer Res ; 59(22): 5719-23, 1999 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-10582690

RESUMEN

We demonstrated previously elevated caveolin-1 expression in metastatic mouse and human prostate cancer cells both in vitro and in vivo. In this study, we analyzed its prognostic value for progression of clinically confined human prostate cancer. Immunohistochemical staining with a caveolin-1-specific antibody was performed on routinely processed paraffin sections from 189 radical prostatectomy specimens. Caveolin-1 immunoreactivity was evaluated in association with patients' age, race, preoperative prostate-specific antigen, clinical stage, and pathological features including Gleason score, extraprostatic extension, status of surgical margins, and time to disease progression after surgery. Positive caveolin-1 immunostaining was detected in 47 of the 189 cancers (25%) and correlated positively with Gleason score, positive surgical margin, as well as lymph node involvement (P = 0.0071, 0.0267, and 0.0399, respectively). In lymph node-negative cancers (n = 162), caveolin-1 immunoreactivity predicts a shorter time to disease progression after surgery (P = 0.0033, univariate analysis). Multivariate analyses that included caveolin-1 and other prognostic pathological markers identified positive caveolin-1 immunostaining as an independent predictor for time to disease progression (P = 0.0186). Thus, our study establishes caveolin-1 as a novel prognostic marker for clinically confined human prostate cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Caveolinas , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Adulto , Anciano , Caveolina 1 , Progresión de la Enfermedad , Humanos , Inmunohistoquímica , Metástasis Linfática , Masculino , Persona de Mediana Edad , Análisis Multivariante , Neoplasia Residual/metabolismo , Pronóstico , Neoplasias de la Próstata/patología , Estudios Retrospectivos
16.
Cancer Res ; 60(20): 5857-61, 2000 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11059783

RESUMEN

Tumor-associated macrophages (TAMs) are highly active immune effector cells that may either positively or negatively regulate the growth of various malignant cells, depending on the biological context. However, the role of TAMs in human prostate cancer progression is unclear. TAMs were immunohistochemically labeled using a monoclonal (CD68) antibody in radical prostatectomy specimens derived from 81 prostate cancer patients. CD68-positive cells were counted with the aid of a microscope and expressed as macrophage index (MphiI), including TAMs/mm2 total tumor tissue (MphiItotal), TAMs/mm2 tumor stroma (MphiIstroma), and TAMs/mm2 cancer cell area (MphiIcancer). MphiIs were analyzed in association with patients' clinical and pathological stage, recurrence status, and histological grade of the cancer. There were significant inverse relationships between MphiItotal and MphiIstroma and clinical stage (P = 0.016 and P = 0.006, respectively). Reduced MphiItotal was also associated with the presence of positive lymph nodes (P = 0.010). Interestingly, although all of the MphiIs differed between Gleason score groups, only MphiIcancer was positively associated with Gleason score. Univariate analysis of MphiItotal and multivariate analysis of MphiItotal with specific pathological markers revealed that MphiItotal was an independent predictor for disease-free survival after surgery (Cox proportional hazard model, P = 0.044 and P = 0.007, respectively). For patients with high MphiItotal (> or = 185.8, the mean MphiItotal value), the disease-free probability 5 years after surgery was 0.75, which was significantly higher than for those with low MphiItotal (0.31, P = 0.0008). Additional immunohistochemical studies that evaluated cytotoxicity-related biomarkers in stroma-associated mononuclear cells suggested reduced functional activities in highly aggressive prostate cancer compared with less aggressive disease. Our results indicate that reduced MphiItotal is a novel prognostic marker for prostate cancer.


Asunto(s)
Macrófagos/inmunología , Neoplasias de la Próstata/inmunología , Anciano , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Humanos , Inmunohistoquímica , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Neoplasias de la Próstata/patología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1 , Factor de Necrosis Tumoral alfa/metabolismo
17.
Cancer Res ; 58(6): 1285-90, 1998 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-9515817

RESUMEN

To identify genes associated with prostate cancer progression, we developed a strategy involving the use of differential display PCR and a panel of genetically matched primary tumor- and metastasis-derived mouse prostate cancer cell lines. We analyzed sequences that were differentially stimulated by transforming growth factor-beta1 in primary tumor-versus metastasis-derived cell lines, based on our previous studies indicating that acquisition of differential responses to this growth factor could result in phenotypic traits that facilitate tumor metastasis from specific cell clones within the primary tumor. Using this system, we isolated and sequenced a cDNA fragment that encoded mouse lysyl oxidase (LO) and was induced by transforming growth factor-beta1 in primary tumor but not in metastasis-derived cells. Northern blotting analysis revealed increased LO expression in a panel of primary tumor cell lines but significantly reduced or nondetectable expression in their matched metastatic counterparts. Further in situ hybridization analysis revealed LO expression in normal mouse prostate epithelium but, in most cases, progressive loss of expression in primary prostate cancer and associated metastatic lesions. Importantly, in situ hybridization studies of normal human prostate and prostate malignancies revealed a similar loss of expression during progression to metastasis. The progressive loss of LO expression during prostate cancer progression provides information that may increase our understanding of the mechanisms that underlie this disease. In addition, LO may provide a useful molecular marker and/or establish a novel therapeutic target for prostate cancer.


Asunto(s)
Neoplasias de la Próstata/enzimología , Proteína-Lisina 6-Oxidasa/genética , Animales , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Hibridación in Situ , Masculino , Ratones , Metástasis de la Neoplasia , ARN Mensajero/genética , ARN Neoplásico/genética , Factor de Crecimiento Transformador beta/farmacología
18.
Cancer Res ; 61(11): 4386-92, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11389065

RESUMEN

Previously, we demonstrated that up-regulation of caveolin-1 (cav-1) was associated with prostate cancer metastasis, biochemical recurrence after radical prostatectomy, and androgen insensitivity. The objective of this study was to characterize the regulation of cav-1 by testosterone (T) and to test the effects of cav-1 on prostate cancer cell survival/clonal growth and metastatic activities. Our results demonstrated that T up-regulated cav-1 protein levels in part through transcriptional regulation and significantly enhanced survival of prostate cancer cell lines ABAC3 and LNCaP after serum starvation (>40% and >60% increased viability, respectively) and in an extended clonogenic assay (approximately 4-fold and 6-fold increase in colonies, respectively). Importantly, antisense cav-1 inhibited the survival effects of T in these assay systems. Modest but not high levels of adenoviral vector-mediated cav-1 expression alone also significantly increased viability (>40%) and clonal growth (10-fold increase in colonies) after serum starvation. Analysis of spontaneous metastasis in stably transfected antisense cav-1 mouse prostate cancer cell clones demonstrated reduction of spontaneous lymph node metastasis incidence (13%), spontaneous lymph node metastasis volume (46%), and experimental lung metastasis incidence (40%) compared with vector control cell clones. Surgical castration further reduced spontaneous lymph node metastasis incidence and volume (18% and 28%, respectively) in antisense cancer cell clones, but not in vector control clones. Our studies demonstrate that cav-1 is a downstream effector of T-mediated prostate cancer cell survival/clonal growth and that modest levels of cav-1 can independently promote prostate cancer cell survival/clonal growth and metastatic activities.


Asunto(s)
Caveolinas/fisiología , Neoplasias Hormono-Dependientes/patología , Neoplasias de la Próstata/patología , Testosterona/farmacología , Animales , Caveolina 1 , Caveolinas/biosíntesis , Caveolinas/genética , División Celular/efectos de los fármacos , División Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , ADN sin Sentido/genética , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Pulmonares/secundario , Metástasis Linfática , Masculino , Ratones , Metástasis de la Neoplasia , Regiones Promotoras Genéticas , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Testosterona/antagonistas & inhibidores , Testosterona/fisiología , Activación Transcripcional/efectos de los fármacos , Células Tumorales Cultivadas , Regulación hacia Arriba
19.
Cancer Res ; 61(10): 3882-5, 2001 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11358800

RESUMEN

Caveolin-1 is an integral protein of caveolae, known to play important roles in signal transduction and lipid transport. We demonstrate that caveolin-1 expression is significantly increased in primary and metastatic human prostate cancer after androgen ablation therapy. We also show that caveolin-1 is secreted by androgen-insensitive prostate cancer cells, and that this secretion is regulated by steroid hormones. Significantly, caveolin-1 was detected in the MDL(3) fraction of serum specimens from patients with advanced prostate cancer and to a lesser extent in normal subjects. Conditioned media from high passage caveolin-1 secreting, androgen-insensitive, LNCaP cells stimulated increased viability and clonal growth of low passage, caveolin-1-negative, androgen-sensitive, LNCaP cells in vitro, and this effect was blocked by treating the media with caveolin-1 antibody. i.p. injections of caveolin-1 antibody suppressed the orthotopic growth and spontaneous metastasis of highly metastatic, androgen-insensitive caveolin-1-secreting mouse prostate cancer. Overall, our results establish caveolin-1 as an autocrine/paracrine factor that is associated with androgen-insensitive prostate cancer. We demonstrate the potential for caveolin-1 as a therapeutic target for this important malignancy.


Asunto(s)
Caveolinas/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Antagonistas de Andrógenos/farmacología , Animales , Anticuerpos/farmacología , Antineoplásicos Hormonales/farmacología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Caveolina 1 , Caveolinas/antagonistas & inhibidores , Caveolinas/biosíntesis , División Celular/fisiología , Supervivencia Celular/fisiología , Medios de Cultivo Condicionados , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología
20.
Cancer Res ; 53(19): 4461-5, 1993 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-8402613

RESUMEN

Several epidemiological studies have implicated low dietary and serum levels of retinol with an increased risk for the development of human prostate cancer. In a recent report, dietary fenretinide [N-[(4-hydroxyphenyl)] retinamide], a synthetic retinoid with low toxicity, decreased the incidence of experimentally induced prostate cancer. Fenretinide is currently being evaluated in phase I and phase II clinical trials as an agent for both the treatment and chemoprevention of human prostate cancer. Because of these findings, we investigated whether dietary fenretinide could alter the incidence of phenotype of oncogene-induced prostate cancer in the mouse prostate reconstitution model system. When compared to control-fed animals, dietary fenretinide reduced the tumor incidence by 49% and the tumor mass by 52% of ras+myc-induced cancers in the mouse prostate reconstitution model system, which was modified to prolong the latency period before cancer development. Retinoids have a wide ranging effect on cellular differentiation, growth factor synthesis, and immune function. While its mechanism of action in this system remains unclear, fenretinide is an effective agent for the chemoprevention and growth modulation of oncogene-induced prostate cancer in the mouse prostate reconstitution model system and may be effective for the chemoprevention of human prostate cancer.


Asunto(s)
Anticarcinógenos/uso terapéutico , Transformación Celular Neoplásica/efectos de los fármacos , Fenretinida/uso terapéutico , Genes myc , Genes ras , Próstata/patología , Neoplasias de la Próstata/prevención & control , Animales , Anticarcinógenos/administración & dosificación , Anticarcinógenos/farmacología , Dieta , Fenretinida/administración & dosificación , Fenretinida/farmacología , Feto , Vectores Genéticos , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Próstata/efectos de los fármacos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transfección
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda