Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Oncotarget ; 4(12): 2366-82, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24280306

RESUMO

Spheroid formation is one property of stem cells-such as embryo-derived or neural stem cells-that has been used for the enrichment of cancer stem-like cells (CSLCs). However, it is unclear whether CSLC-derived spheroids are heterogeneous or whether they share common embryonic stemness properties. Understanding these features might lead to novel therapeutic approaches. Ovarian carcinoma is a deadly disease of women. We identified two types of spheroids (SR1 and SR2) from ovarian cancer cell lines and patients' specimens according to their morphology. Both types expressed stemness markers and could self-renew and initiate tumors when a low number of cells were used. Only SR1 could differentiate into multiple-lineage cell types under specific induction conditions. SR1 spheroids could differentiate to SR2 spheroids through epithelial-mesenchymal transition. Alkaline phosphatase (ALP) was highly expressed in SR1 spheroids, decreased in SR2 spheroids, and was absent in differentiated progenies in accordance with the loss of stemness properties. We verified that ALP can be a marker for ovarian CSLCs, and patients with greater ALP expression is related to advanced clinical stages and have a higher risk of recurrence and lower survival rate. The ALP inhibitor, levamisole, disrupted the self-renewal of ovarian CSLCs in vitro and tumor growth in vivo. In summary, this research provides a plastic ovarian cancer stem cell model and a new understanding of the cross-link between stem cells and cancers.This results show that ovarian CSLCs can be suppressed by levamisole. Our findings demonstrated that some ovarian CSLCs may restore ALP activity, and this suggests that inhibition of ALP activity may present a new opportunity for treatment of ovarian cancer.


Assuntos
Fosfatase Alcalina/antagonistas & inibidores , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neoplasias Epiteliais e Glandulares/enzimologia , Células-Tronco Neoplásicas/patologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Adolescente , Adulto , Idoso , Fosfatase Alcalina/metabolismo , Carcinoma Epitelial do Ovário , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Linhagem da Célula , Transição Epitelial-Mesenquimal , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/enzimologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Esferoides Celulares , Adulto Jovem
2.
PLoS One ; 8(9): e74538, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24058587

RESUMO

The primary cause of death from breast cancer is the progressive growth of tumors and resistance to conventional therapies. It is currently believed that recurrent cancer is repopulated according to a recently proposed cancer stem cell hypothesis. New therapeutic strategies that specifically target cancer stem-like cells may represent a new avenue of cancer therapy. We aimed to discover novel compounds that target breast cancer stem-like cells. We used a dye-exclusion method to isolate side population (SP) cancer cells and, subsequently, subjected these SP cells to a sphere formation assay to generate SP spheres (SPS) from breast cancer cell lines. Surface markers, stemness genes, and tumorigenicity were used to test stem properties. We performed a high-throughput drug screening using these SPS. The effects of candidate compounds were assessed in vitro and in vivo. We successfully generated breast cancer SPS with stem-like properties. These SPS were enriched for CD44(high) (2.8-fold) and CD24(low) (4-fold) cells. OCT4 and ABCG2 were overexpressed in SPS. Moreover, SPS grew tumors at a density of 10(3), whereas an equivalent number of parental cells did not initiate tumor formation. A clinically approved drug, niclosamide, was identified from the LOPAC chemical library of 1,258 compounds. Niclosamide downregulated stem pathways, inhibited the formation of spheroids, and induced apoptosis in breast cancer SPS. Animal studies also confirmed this therapeutic effect. The results of this proof-of-principle study may facilitate the development of new breast cancer therapies in the near future. The extension of niclosamide clinical trials is warranted.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Células-Tronco Neoplásicas/patologia , Niclosamida/uso terapêutico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Separação Celular , Avaliação Pré-Clínica de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Camundongos SCID , Células-Tronco Neoplásicas/efeitos dos fármacos , Niclosamida/farmacologia , Células da Side Population/efeitos dos fármacos , Células da Side Population/metabolismo , Células da Side Population/patologia , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia
3.
Gynecol Oncol ; 128(3): 475-82, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23270808

RESUMO

OBJECTIVE: We reported recently the hypermethylation of LMX1A, a LIM-homeobox gene, as a prognostic biomarker in ovarian cancer; however, the function of LMX1A in ovarian cancer remains unknown. The present study aimed to evaluate the hypothesized tumour-suppressor functions of LMX1A in ovarian cancer. METHODS: We analysed the function of LMX1A by examining cell lines, animal models and human ovarian cancer tissues. Overexpression of LMX1A in relation to chemotherapy was also analysed. RESULTS: The expression of LMX1A inhibited cell proliferation, migration, invasion and colony formation in vitro, as well as tumourigenicity in a xenotransplantation mouse model. LMX1A also sensitized ovarian cancer cell lines to chemotherapeutics, and affected epithelial-mesenchymal transition (EMT). The restoration of LMX1A down-regulated stem cell markers and inhibited tumour spheroid formation in SKOV3 cells. Univariate analysis of immunohistochemical staining of tissue arrays (n=83) revealed that low LMX1A expression was significantly associated with advanced stages (p=0.001), poor differentiation (p<0.001), early recurrence (p=0.023) and poor overall survival (p=0.042) in ovarian cancer. CONCLUSIONS: The present study demonstrated, for the first time, that LMX1A is a bona fide tumour suppressor of ovarian cancer. The prognostic values of LMX1A may provide a biomarker for personalized treatments of ovarian cancer patients. The mechanisms of LMX1A in EMT and stem-like properties in ovarian cancer warrant further investigation.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Genes Supressores de Tumor , Proteínas com Homeodomínio LIM/genética , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Fatores de Transcrição/genética , Animais , Carcinoma Epitelial do Ovário , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Metilação de DNA , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Feminino , Humanos , Proteínas com Homeodomínio LIM/biossíntese , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Neoplasias Epiteliais e Glandulares/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias Ovarianas/metabolismo , Fatores de Transcrição/biossíntese , Transfecção
4.
PLoS One ; 7(7): e41060, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22815913

RESUMO

BACKGROUND: Despite of the trend that the application of DNA methylation as a biomarker for cancer detection is promising, clinically applicable genes are few. Therefore, we looked for novel hypermethylated genes for cervical cancer screening. METHODS AND FINDINGS: At the discovery phase, we analyzed the methylation profiles of human cervical carcinomas and normal cervixes by methylated DNA immunoprecipitation coupled to promoter tiling arrays (MeDIP-on-chip). Methylation-specific PCR (MSP), quantitative MSP and bisulfite sequencing were used to verify the methylation status in cancer tissues and cervical scrapings from patients with different severities. Immunohistochemical staining of a cervical tissue microarray was used to confirm protein expression. We narrowed to three candidate genes: DBC1, PDE8B, and ZNF582; their methylation frequencies in tumors were 93%, 29%, and 100%, respectively. At the pre-validation phase, the methylation frequency of DBC1 and ZNF582 in cervical scraping correlated significantly with disease severity in an independent cohort (n = 330, both P<0.001). For the detection of cervical intraepithelial neoplasia 3 (CIN3) and worse, the area under the receiver operating characteristic curve (AUC) of ZNF582 was 0.82 (95% confidence interval= 0.76-0.87). CONCLUSIONS: Our study shows ZNF582 is frequently methylated in CIN3 and worse lesions, and it is demonstrated as a potential biomarker for the molecular screening of cervical cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição Kruppel-Like/biossíntese , Fatores de Transcrição Kruppel-Like/genética , Neoplasias do Colo do Útero/metabolismo , Dedos de Zinco , 3',5'-AMP Cíclico Fosfodiesterases/biossíntese , 3',5'-AMP Cíclico Fosfodiesterases/genética , Biomarcadores/metabolismo , Proteínas de Ciclo Celular , Ilhas de CpG , Metilação de DNA , Feminino , Células HeLa , Humanos , Proteínas do Tecido Nervoso , Reação em Cadeia da Polimerase/métodos , Regiões Promotoras Genéticas , Curva ROC , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética
5.
Mol Cancer Ther ; 11(8): 1703-12, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22576131

RESUMO

A recent hypothesis for cancer chemoresistance posits that cytotoxic survival of a subpopulation of tumor progenitors drives the propagation of recurrent disease, underscoring the need for new therapeutics that target such primitive cells. To discover such novel compounds active against drug-resistant ovarian cancer, we identified a subset of chemoresistant ovarian tumor cells fulfilling current definitions of cancer-initiating cells from cell lines and patient tumors using multiple stemness phenotypes, including the expression of stem cell markers, membrane dye efflux, sphere formation, potent tumorigenicity, and serial tumor propagation. We then subjected such stem-like ovarian tumor-initiating cells (OTIC) to high-throughput drug screening using more than 1,200 clinically approved drugs. Of 61 potential compounds preliminarily identified, more stringent assessments showed that the antihelmintic niclosamide selectively targets OTICs in vitro and in vivo. Gene expression arrays following OTIC treatment revealed niclosamide to disrupt multiple metabolic pathways affecting biogenetics, biogenesis, and redox regulation. These studies support niclosamide as a promising therapy for ovarian cancer and warrant further preclinical and clinical evaluation of this safe, clinically proven drug for the management of this devastating gynecologic malignancy.


Assuntos
Antineoplásicos/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Niclosamida/farmacologia , Neoplasias Ovarianas/metabolismo , Animais , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Análise por Conglomerados , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Niclosamida/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Células da Side Population/efeitos dos fármacos , Células da Side Population/metabolismo , Bibliotecas de Moléculas Pequenas , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Am J Pathol ; 180(3): 1159-1169, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22222226

RESUMO

The role of aldehyde dehydrogenase 1 (ALDH1) as an ovarian cancer stem cell marker and its clinical significance have rarely been explored. We used an Aldefluor assay to isolate ALDH1-bright (ALDH1(br)) cells from epithelial ovarian cancer cell lines and characterized the properties of the stem cells. ALDH1(br) cells were enriched in ES-2 (1.3%), TOV-21G (1.0%), and CP70 (1.2%) cells. Both ALDH1(br) and ALDH1(low) cells repopulated stem cell heterogeneity, formed spheroids, and grew into tumors in immunocompromised mice, although these processes were more efficient in ALDH1(br) cells. In the ES-2 and CP70 cells, ALDH1(br) cells conferred more chemoresistance, and were more enriched in CD44 (by 1.74-fold and 5.18-fold, respectively) than in CD133 (by 1.39-fold and 1.17-fold, respectively), compared with ALDH1(low) cells. Immunohistochemical staining for ALDH1 on a tissue microarray containing 84 epithelial ovarian cancer samples revealed that patients with higher ALDH1 expression (>50%) had poor overall survival, compared with those with lower ALDH1 (P = 0.004) and yielded an odds ratio of death of 2.43 (95% CI = 1.12 to 5.28) by multivariate analysis. The results did not support ALDH1 alone as an ovarian cancer stem cell marker, but demonstrated that ALDH1 is associated with CD44 expression, chemoresistance, and poor clinical outcome. The use of a combination of ALDH1 with other stem cell markers may help define ovarian cancer stem cells more stringently.


Assuntos
Biomarcadores Tumorais/metabolismo , Receptores de Hialuronatos/metabolismo , Isoenzimas/metabolismo , Neoplasias Epiteliais e Glandulares/enzimologia , Neoplasias Ovarianas/metabolismo , Retinal Desidrogenase/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Família Aldeído Desidrogenase 1 , Animais , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/enzimologia , Recidiva Local de Neoplasia/mortalidade , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neoplasias Epiteliais e Glandulares/mortalidade , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/mortalidade , Esferoides Celulares , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
7.
Hum Gene Ther ; 22(2): 145-54, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20825285

RESUMO

Indoleamine 2,3-dioxygenase (IDO) has been known as an emerging therapeutic target in autoimmunity-related arthritis. The treatment responses of adenoviral vectors encoding IDO (AdIDO) gene therapy in rat collagen-induced arthritis (CIA) were examined in this study. The therapeutic effects on ankle circumference, articular index, and radiographic and histological scores were evaluated in AdIDO-injected ankle joints. We further determined CD4+ T-cell numbers and their apoptotic status, CD68(+) macrophage numbers, kynurenine (a downstream tryptophan metabolite) concentrations, interleukin-17 (IL-17) levels, and retinoic acid-related orphan receptor γt (RORγt) expression in synovial tissues of CIA rats receiving AdIDO treatment. Reduction of ankle circumference, articular index, and radiographic and histological scores were noted in AdIDO-treated ankles, as compared with those receiving injection of control vectors. Furthermore, IDO gene transfer led to decreased infiltrating CD4+ T cells with enhanced apoptosis, reduced CD68+ macrophage numbers, increased kynurenine levels, lower IL-17 concentrations, and decreased RORγt expression within the ankle joints. In addition, such a therapy diminished type II collagen-specific IL-17 production and RORγt expression in CD4+ T cells from draining lymph nodes of CIA rats. Our results demonstrate for the first time that intra-articular delivery of IDO gene ameliorated ankle arthritis of CIA rats by induction of CD4+ T-cell apoptosis and reduction of synovial IL-17 production through the supplement of kynurenine. Taken together, these findings implicate the novel strategy of using IDO gene as a therapeutic approach in treating patients with rheumatoid arthritis.


Assuntos
Apoptose , Artrite Experimental/terapia , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interleucina-17/análise , Adenoviridae/genética , Animais , Articulação do Tornozelo/metabolismo , Artrite Reumatoide/terapia , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Proliferação de Células , Terapia Genética , Vetores Genéticos , Humanos , Cinurenina/análise , Macrófagos/imunologia , Ratos , Ratos Sprague-Dawley , Membrana Sinovial/metabolismo , Triptofano/análise
8.
J Agric Food Chem ; 57(18): 8266-73, 2009 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-19711916

RESUMO

Licorice is a common Chinese medicinal herb with antitumor activity. Some components in licorice root have been shown to induce cell cycle arrest or apoptosis in cancer cells. This paper demonstrates for the first time that licorice Glycyrrhiza glabra and its component licochalcone-A (LA) can induce autophagy in addition to apoptosis in human LNCaP prostate cancer cells. Exposure of cells to licorice or LA resulted in several confirmed characteristics of autophagy, including the appearance of autophagic vacuoles revealed by monodansylcadaverine (MDC) staining, formation of acidic vesicular organelles (AVOs), and autophagosome membrane association of microtubule-associated protein 1 light chain 3 (LC3) characterized by cleavage of LC3 and its punctuate redistribution, as well as ultrastructural observation of autophagic vacuoles by transmission electron microscopy. Autophagy induction was accompanied by down-regulation of Bcl-2 and inhibition of the mammalian target of rapamycin (mTOR) pathway. In summary, licorice can induce caspase-dependent and autophagy-related cell death in LNCaP cells.


Assuntos
Autofagia/efeitos dos fármacos , Chalconas/farmacologia , Genes bcl-2/efeitos dos fármacos , Glycyrrhiza/química , Neoplasias da Próstata/ultraestrutura , Proteínas Quinases/efeitos dos fármacos , Linhagem Celular Tumoral , Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Microscopia Eletrônica de Transmissão , Neoplasias da Próstata/metabolismo , Serina-Treonina Quinases TOR , Vacúolos/ultraestrutura
9.
Autophagy ; 5(4): 451-60, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19164894

RESUMO

Cathepsins have long been considered as housekeeping molecules. However, specific functions have also been attributed to each of these lysosomal proteases. Squamous cell carcinoma antigen (SCCA) 1, widely expressed in various uterine cervical cells, is an endogenous cathepsin (cat) L inhibitor. In this study, we investigated whether the cat L-SCCA 1 lysosomal pathway and autophagy were involved in resveratrol (RSV)-induced cytotoxicity in cervical cancer cells. RSV induced GFP-LC3 aggregation as well as increased the presence of LC3-II and autophagosomes as was revealed by electron microscopy in cervical cancer cells. Prolonged treatment of RSV induced cytosolic translocation of cytochrome c, caspase 3 activation and apoptotic cell death. This apoptotic effect was abrogated by trans-epoxysuccinyl-L-leucylamido-(4-guanidino)butane, an inhibitor of cat B and L, but not by pepstatin A, an inhibitor of cat D. As cervical cancer cells express little cat B, we further studied the role of cat L. RSV induced dissipation of the lysosomal membrane permeability (LMP), leakage and increased cytosolic expression and activity of cat L. Inhibition of cat L by small interference RNA (siRNA) protected cells from RSV-induced cytotoxicity. In contrast, inhibition of SCCA 1 by siRNA promoted RSV-induced cytotoxicity. Inhibition of autophagic response by wortmannin (WT) or asparagine (ASP) resulted in decreased early LC3-II formation, reduced LMP, and abolishment of the increase in RSV-induced cell death. In conclusion, we have identified a new cytotoxic mechanism in which the lysosomal enzyme cat L acts as a death signal integrator in cervical cancer cells. Furthermore, SCCA 1 may play an antiapoptotic role through anti-cat L activity.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Catepsinas/metabolismo , Cisteína Endopeptidases/metabolismo , Estilbenos/farmacologia , Neoplasias do Colo do Útero/enzimologia , Neoplasias do Colo do Útero/patologia , Caspase 3/metabolismo , Catepsina L , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citocromos c/metabolismo , Citosol/efeitos dos fármacos , Citosol/enzimologia , Ativação Enzimática/efeitos dos fármacos , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Feminino , Humanos , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/ultraestrutura , Ferro/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Modelos Biológicos , Permeabilidade/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Resveratrol , Neoplasias do Colo do Útero/ultraestrutura
10.
Arthritis Rheum ; 58(6): 1650-6, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18512785

RESUMO

OBJECTIVE: The phosphatidylinositol 3-kinase (PI 3-kinase)/Akt pathway is known to be activated in rheumatoid arthritis (RA) synovial tissue, which impacts cell growth, proliferation, survival, and migration. Phosphatase and tensin homolog deleted from chromosome 10 (PTEN) functions as a negative regulator of PI 3-kinase signaling, thus blocking Akt activation. The aim of this study was to examine the effect of PTEN gene transfer in rats with collagen-induced arthritis (CIA). METHODS: Adenoviral vectors encoding human PTEN (AdPTEN) or beta-galactosidase (AdLacZ) were injected intraarticularly into rats with CIA, and their treatment responses were monitored by measures of clinical, radiographic, and histologic changes. The expression of phosphorylated Akt, total Akt, vascular endothelial growth factor (VEGF), proinflammatory cytokines, and chemokines, as well as the extent of microvessel density in the ankle joints were determined. RESULTS: AdPTEN treatment reduced Akt phosphorylation and decreased VEGF production in human RA synovial fibroblasts. Compared with AdLacZ treatment of the rats with CIA, AdPTEN treatment significantly reduced ankle circumference, articular index scores, radiography scores, and histology scores, and also decreased microvessel density and levels of VEGF and interleukin-1beta. Furthermore, PTEN gene transfer led to down-regulation of Akt activation and increased apoptosis in the ankle joints. CONCLUSION: This study is the first to demonstrate the in vivo effect of intraarticular gene delivery of PTEN on amelioration of arthritis symptoms in rats with CIA, which involved antiangiogenic, antiproliferative, and antiinflammatory effects of PTEN via inhibition of the PI 3-kinase/Akt signaling pathway. Our findings also implicate the PI 3-kinase/Akt pathway as a therapeutic target for the treatment of RA or other inflammatory diseases.


Assuntos
Artrite Experimental/terapia , Fibroblastos/metabolismo , Terapia Genética/métodos , PTEN Fosfo-Hidrolase/genética , Adenoviridae/genética , Animais , Articulação do Tornozelo/patologia , Artrite Experimental/patologia , Células Cultivadas , Transferência Genética Horizontal , Vetores Genéticos , Humanos , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
11.
Clin Cancer Res ; 14(4): 1228-38, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18281558

RESUMO

PURPOSE: Oncolytic adenoviruses are attractive therapeutics for cancer because they selectively replicate in tumors. However, targeting tumor metastasis remains a major challenge for current virotherapy for cancer. Oct-3/4 is specifically expressed in embryonic stem cells and tumor cells. Oct-3/4 highly expressed in cancer cells may be a potential target for cancer therapy. We developed an E1B-55 kDa-deleted adenovirus, designated Ad.9OC, driven by nine copies of Oct-3/4 response element for treating Oct-3/4-expressing metastatic bladder cancer. EXPERIMENTAL DESIGN: We examined the expression of Oct-3/4 in human bladder tumor tissues and bladder cancer cell lines. We also evaluated the cytolytic and antitumor effects of Ad.9OC on bladder cancer cells in vitro and in vivo. RESULTS: Oct-3/4 expression was detected in bladder cancer cell lines, as well as in human bladder tumor tissues. Notably, Oct-3/4 expression was higher in metastatic compared with nonmetastatic bladder cancer cells. Ad.9OC induced higher cytolytic activity in metastatic bladder cancer cells than in their nonmetastatic counterparts, whereas it did not cause cytotoxicity in normal cells. Pharmacologic and short hairpin RNA-mediated Oct-3/4 inhibition rendered bladder cancer cells more resistant to Ad.9OC-induced cytolysis. Replication of Ad.9OC was detected in murine bladder cancer cells and bladder tumor tissues. We also showed the effectiveness of Ad.9OC for treating bladder cancer in subcutaneous, as well as metastatic, bladder tumor models. CONCLUSIONS: Ad.9OC may have therapeutic potential for treating Oct-3/4-expressing tumors. Especially, metastatic bladder tumors are good target for Ad.9OC treatment. Because Oct-3/4 is expressed in a broad spectrum of cancers, Ad.9OC may be broadly applicable.


Assuntos
Fator 3 de Transcrição de Octâmero/metabolismo , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Neoplasias da Bexiga Urinária/terapia , Neoplasias da Bexiga Urinária/virologia , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Immunoblotting , Imuno-Histoquímica , Camundongos , Fator 3 de Transcrição de Octâmero/genética , Elementos de Resposta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ativação Transcricional , Ensaios Antitumorais Modelo de Xenoenxerto
12.
J Immunol ; 178(7): 4688-94, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17372028

RESUMO

Prothymosin alpha (ProT) is regulated by c-Myc, an oncoprotein overexpressed in synovium of rheumatoid arthritis, and is associated with cell proliferation. However, ProT also exerts immunomodulatory activities. The growth-promoting activity of ProT can be abolished by deleting its nuclear localization signal (NLS). In this study, we showed that AdProTDeltaNLS, an adenoviral vector encoding ProT lacking the NLS, did not enhance the proliferation of synovial fibroblasts. AdProTDeltaNLS treatment abolished the up-regulation of the MIP-1alpha promoter activity induced by TNF-alpha in synovial fibroblasts. AdProTDeltaNLS suppressed macrophage chemotaxis and reduced macrophage infiltration into the ankle joints in rats with collagen-induced arthritis (CIA). Neutralization test confirmed the involvement of MIP-1alpha in macrophage chemotaxis. Administration of AdProTDeltaNLS reduced the severity of CIA in the clinical, radiographic, and histological aspects. The levels of TNF-alpha (mean +/- SEM, 1261.9 +/- 107.9 vs 2880.1 +/- 561.4 pg/mg total protein; p < 0.05), IL-1beta (56.8 +/- 8.0 vs 109.2 +/- 4.9 pg/mg total protein; p < 0.01), and MIP-1alpha (41.7 +/- 3.6 vs 55.2 +/- 1.1 pg/mg total protein; p < 0.05) in the ankle joints were lower in the AdProTDeltaNLS-treated rats with CIA than those in their control counterparts. In the AdProTDeltaNLS-treated ankle joints, matrix metalloproteinase-9 expression was decreased by 40% and infiltrating macrophages reduced by 50%. Our results demonstrate that intra-articular delivery of AdProTDeltaNLS significantly ameliorated the clinical course of CIA in rats. This study is the first to suggest that ProT lacking the NLS may have therapeutic potential for the management of rheumatoid arthritis.


Assuntos
Artrite Reumatoide/terapia , Terapia Genética , Vetores Genéticos , Sinais de Localização Nuclear/genética , Precursores de Proteínas/genética , Timosina/análogos & derivados , Adenoviridae/genética , Animais , Articulação do Tornozelo/imunologia , Articulação do Tornozelo/patologia , Artrite Reumatoide/induzido quimicamente , Artrite Reumatoide/patologia , Proliferação de Células , Quimiocina CCL3 , Quimiocina CCL4 , Quimiotaxia , Colágeno/toxicidade , Fibroblastos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Interleucina-1beta/metabolismo , Proteínas Inflamatórias de Macrófagos/metabolismo , Macrófagos/imunologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Sinais de Localização Nuclear/análise , Precursores de Proteínas/análise , Ratos , Deleção de Sequência , Líquido Sinovial/química , Timosina/análise , Timosina/genética , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
13.
J Cell Biochem ; 100(4): 981-90, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17131359

RESUMO

Ribosomal biogenesis is correlated with cell cycle, cell proliferation, cell growth and tumorigenesis. Some oncogenes and tumor suppressors are involved in regulating the formation of mature ribosome and affecting the ribosomal biogenesis. In previous studies, the mitochondrial ribosomal protein L41 was reported to be involved in cell proliferation regulating through p21(WAF1/CIP1) and p53 pathway. In this report, we have identified a mitochondrial ribosomal protein S36 (mMRPS36), which is localized in the mitochondria, and demonstrated that overexpression of mMRPS36 in cells retards the cell proliferation and delays cell cycle progression. In addition, the mMRPS36 overexpression induces p21(WAF1/CIP1) expression, and regulates the expression and phosphorylation of p53. Our result also indicate that overexpression of mMRPS36 affects the mitochondrial function. These results suggest that mMRPS36 plays an important role in mitochondrial ribosomal biogenesis, which may cause nucleolar stress, thereby leading to cell cycle delay.


Assuntos
Ciclo Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas Mitocondriais/fisiologia , Proteínas Ribossômicas/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Ciclo Celular/genética , Proliferação de Células , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Expressão Gênica , Imuno-Histoquímica , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Microscopia Confocal , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Dados de Sequência Molecular , Células NIH 3T3 , Espécies Reativas de Oxigênio/metabolismo , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Alinhamento de Sequência , Proteína Supressora de Tumor p53/genética
14.
Cancer Res ; 66(20): 9957-66, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17047058

RESUMO

The human telomerase reverse transcriptase (hTERT) promoter can selectively drive transgene expression in many telomerase-positive human cancer cells. Here we evaluated combination therapy of adenoviral vector Ad-hTERT-CD encoding E. coli cytosine deaminase (CD) driven by the hTERT promoter and low-dose etoposide (0.1 microg/mL) for treating bladder cancer. Ad-hTERT-CD conferred sensitivity to 5-fluorocytosine (5-FC) in bladder cancer cells, which could be enhanced by etoposide treatment, but not in normal cells. Such effect was correlated with up-regulation of hypoxia-inducible factor (HIF)-1alpha expression. By contrast, etoposide activated p53 and down-regulated hTERT promoter activity in normal cells. Etoposide also increased adenoviral infection via enhancement of coxsackie-adenovirus receptor expression on bladder cancer and normal cells. Combination index analysis revealed that combined therapy of Ad-hTERT-CD (10(9) plaque-forming units)/5-FC (200 mg/kg) with etoposide (2 mg/kg) synergistically suppressed tumor growth and prolonged survival in mice bearing syngeneic MBT-2 bladder tumors. This combination therapy regimen induced complete tumor regression and generated antitumor immunity in 75% of tumor-bearing mice. Furthermore, increased infiltrating CD4(+) and CD8(+) T cells and necrosis within tumors were found in mice receiving combination therapy of Ad-hTERT-CD and etoposide compared with those treated with either treatment alone. Thus, the potential high therapeutic index of the combination therapy may be an appealing therapeutic intervention for bladder cancer. Furthermore, because a majority of human tumors exhibit high telomerase activity, adenovirus-mediated CD gene therapy driven by the hTERT promoter in combination with low-dose etoposide may be applicable to a broad spectrum of cancers.


Assuntos
Citosina Desaminase/genética , Etoposídeo/farmacologia , Terapia Genética/métodos , Telomerase/genética , Neoplasias da Bexiga Urinária/terapia , Adenoviridae/genética , Animais , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Terapia Combinada , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Citosina Desaminase/biossíntese , Citosina Desaminase/metabolismo , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Flucitosina/farmacologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Endogâmicos C3H , Regiões Promotoras Genéticas , Receptores Virais/biossíntese , Receptores Virais/genética , Telomerase/biossíntese , Transgenes , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Kidney Int ; 67(5): 1710-22, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15840017

RESUMO

BACKGROUND: Polycystic kidney disease (PKD) is a genetic disorder characterized by development of renal cysts and progressive renal dysfunction. Renal tissues from both PKD patients and rodent models of PKD show elevated c-myc expression. Prothymosin alpha (ProT) is positively regulated by c-myc through binding to the E box of its promoter. Through creating transgenic mice and clinical studies, we sought to investigate whether ProT overexpression contributes to PKD development. METHODS: ProT heterozygous and homozygous transgenic mice were generated and characterized. Morphologic, histologic, immunohistochemical, and biochemical analyses of the transgenic mice were performed. RESULTS: Two transgenic lines that represented integration at two different loci of the chromosomes were generated. ProT overexpression in the kidneys of homozygous transgenic mice induced a PKD phenotype, which included polycystic kidneys, elevated blood urea nitrogen (BUN), and lethality at about 10 days of age. Similar overexpression pattern of ProT was noted in cystic kidneys of the transgenic mice as well as in human autosomal-recessive PKD (ARPKD) and autosomal-dominant PKD (ADPKD) kidneys. ProT protein levels in the kidneys and urine as well as renal mRNA level of epithelial growth factor receptor (EGFR) of homozygous ProT transgenic mice were significantly higher than heterozygous or nontransgenic littermates. Furthermore, the heterozygous transgenic mice at 17 months of age also developed mild cystic kidneys. CONCLUSION: Transgenic mice overexpressing ProT represent a novel model for PKD and may provide insights into PKD development. ProT, like c-myc and EGFR, may contribute to the development of renal cysts and may be a potential noninvasive diagnostic molecule of PKD.


Assuntos
Doenças Renais Policísticas/etiologia , Doenças Renais Policísticas/genética , Precursores de Proteínas/genética , Timosina/análogos & derivados , Timosina/genética , Animais , Sequência de Bases , Nitrogênio da Ureia Sanguínea , DNA Complementar/genética , Modelos Animais de Doenças , Receptores ErbB/genética , Expressão Gênica , Humanos , Rim/imunologia , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Transgênicos , Fenótipo , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/patologia , Precursores de Proteínas/metabolismo , Timosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA