Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Am Anim Hosp Assoc ; 50(2): 96-104, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24446402

RESUMO

An image-guided robotic stereotactic radiosurgery (SRS) system can be used to deliver curative-intent radiation in either single fraction or hypofractionated doses. Medical records for 19 dogs with nonlymphomatous nasal tumors treated with hypofractionated image-guided robotic stereotactic body radiotherapy (SBRT), either with or without adjunctive treatment, were retrospectively analyzed for survival and prognostic factors. Median survival time (MST) was evaluated using Kaplan-Meier survival curves. Age, breed, tumor type, stage, tumor size, prescribed radiation dose, and heterogeneity index were analyzed for prognostic significance. Dogs were treated with three consecutive-day, 8-12 gray (Gy) fractions of image-guided robotic SBRT. Overall MST was 399 days. No significant prognostic factors were identified. Acute side effects were rare and mild. Late side effects included one dog with an oronasal fistula and six dogs with seizures. In three of six dogs, seizures were a presenting complaint prior to SBRT. The cause of seizures in the remaining three dogs could not be definitively determined due to lack of follow-up computed tomography (CT) imaging. The seizures could have been related to either progression of disease or late radiation effect. Results indicate that image-guided robotic SBRT, either with or without adjunctive therapy, for canine nonlymphomatous nasal tumors provides comparable survival times (STs) to daily fractionated megavoltage radiation with fewer required fractions and fewer acute side effects.


Assuntos
Doenças do Cão/radioterapia , Neoplasias Nasais/veterinária , Radioterapia Guiada por Imagem/veterinária , Animais , Doenças do Cão/mortalidade , Doenças do Cão/patologia , Cães , Fracionamento da Dose de Radiação , Metástase Linfática , Neoplasias Nasais/radioterapia , Estudos Retrospectivos , Análise de Sobrevida , Resultado do Tratamento
2.
J Urol ; 190(4): 1404-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23500642

RESUMO

PURPOSE: KU7 is a popular urothelial carcinoma cell line that was isolated from the bladder of a patient at Keio University in 1980. It has subsequently been widely used in laboratories around the world. We describe how routine cell line authentication revealed that KU7 was cross contaminated almost 30 years ago with HeLa, a cervical carcinoma cell line. MATERIALS AND METHODS: Presumed KU7 clones dating from 1984 to 1999 were provided by M.D. Anderson Cancer Center, Vancouver Prostate Centre, Kyoto University, Tokyo Medical University and Keio University. HeLa was obtained from ATCC. Genomic DNA was isolated and short tandem repeat analysis was performed at the M.D. Anderson Cancer Center Characterized Cell Line Core Facility, Johns Hopkins University Fragment Analysis Facility and RIKEN BioResource Center, Ibaraki, Japan. Comparative genomic hybridization was performed on a platform (Agilent Technologies, Santa Clara, California) at Vancouver Prostate Centre. RESULTS: The short tandem repeat profile of all KU7 clones was an exact match with that of HeLa. Comparative genomic hybridization of all samples revealed an abundance of shared chromosomal aberrations. Slight differences in some genomic areas were explained by genomic drift in different KU7 clones separated by many years. CONCLUSIONS: Our analysis identified that cross contamination of KU7 with HeLa occurred before 1984 at the source institution. All KU7 clones in the urological literature should be considered HeLa and experimental results should be viewed in this light. Our results emphasize the need to authenticate cell lines in oncological research.


Assuntos
Contaminação por DNA , Células HeLa , Hibridização Genômica Comparativa , Perfilação da Expressão Gênica , Humanos , Fatores de Tempo , Neoplasias da Bexiga Urinária/patologia
3.
Curr Nutr Rep ; 11(4): 643-652, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36018501

RESUMO

PURPOSE OF REVIEW: An epidemic of age-associated cognitive decline, most commonly ascribed to neurodegenerative conditions such as Alzheimer's and Parkinson's disease, is causing healthcare costs to soar and devastating caregivers. An estimated 6.5 million Americans are living today with Alzheimer's disease, with 13.8 million cases projected by mid-century. Although genetic mutations are known to cause neurodegeneration, autosomal dominant disease is very rare and most sporadic cases can be attributed, at least in part, to modifiable risk factors. RECENT FINDINGS: Diet is a potential modifiable risk factor in cognitive decline. Food communicates with the brain through a complex signaling web involving multiple cells, mediators and receptors. Gut-brain communication is modulated by microorganisms including bacteria, archaea, viruses, and unicellular eukaryotes, which together constitute the microbiota. Microbes not only play major roles in the digestion and fermentation of the food, providing nutrients and bioactive metabolites, but also reflect the type and amount of food consumed and food-borne toxic exposures. Food components modify the diversity and abundance of the microbial populations, maintain the integrity of the gut barrier, and regulate the passage of microbes and their metabolites into the blood stream where they modulate the immune system and communicate with body systems including the brain. This paper will focus on selected mechanisms through which interactions between diet and the gut microbiota can modify brain integrity and cognitive function with emphasis on the pathogenesis of the most common dementia, Alzheimer's disease.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Microbioma Gastrointestinal , Humanos , Microbioma Gastrointestinal/fisiologia , Dieta , Encéfalo/metabolismo
4.
Prostate ; 70(3): 239-51, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19790237

RESUMO

BACKGROUND: De novo androgen synthesis and subsequent androgen receptor (AR) activation has recently been shown to contribute to castration-resistant prostate cancer (CRPC) progression. Herein we provide evidence that fatty acids (FA) can trigger androgen synthesis within steroid starved prostate cancer (CaP) tumor cells. METHODS: Tumoral FA and steroid levels were assessed by GC-MS and LC-MS, respectively. Profiles of genes and proteins involved in FA activation of steroidogenesis were assessed by fluorescence microscopy, immunohistochemistry, microarray expression profiling and Western blot analysis. RESULTS: In human CaP tissues the levels of proteins responsible for FA activation of steroid synthesis were observed to be altered during progression to CRPC. Further investigating this mechanism in LNCaP cells, we demonstrate that specific FA, arachidonic acid, is synthesized in an androgen-dependent and AR-mediated manner. Arachidonic acid is known to induce steroidogenic acute regulatory protein (StAR) in steroidogenic cells. When bound to hormone sensitive lipase (HSL), StAR shuttles free cholesterol into the mitochondria for downstream conversion into androgens. We show that arachidonic acid induces androgen production in steroid starved LNCaP cells coincidently in the same conditions that HSL and StAR are predominantly localized in the mitochondria. Furthermore, their activities are verified by a functional increase in mitochondrial uptake of cholesterol in this steroid starved environment. CONCLUSIONS: We propose that this characterized arachidonic acid induced steroidogenesis mechanism significantly contributes to the activation of AR in CRPC progression and therefore recommend that fatty acid pathways be targeted therapeutically in progressing CaP.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Antineoplásicos Hormonais/uso terapêutico , Ácido Araquidônico/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores Androgênicos/metabolismo , Esteroides/biossíntese , Androgênios/deficiência , Androgênios/metabolismo , Animais , Ácido Araquidônico/farmacologia , Linhagem Celular Tumoral , Colesterol/farmacocinética , Progressão da Doença , Resistência a Medicamentos , Humanos , Masculino , Camundongos , Camundongos Nus , Mitocôndrias/metabolismo , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Fosfoproteínas/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Esterol Esterase/metabolismo , Distribuição Tecidual , Transplante Heterólogo , Regulação para Cima
5.
Clin Cancer Res ; 14(18): 5769-77, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18794086

RESUMO

PURPOSE: GLI transcription factors mediate hedgehog signaling and have been implicated in several human malignancies, including prostate cancer. The objectives of this study were to characterize GLI2 expression levels in human prostate cancer cell lines and tissues to test the effect of antisense oligonucleotide (ASO) targeting GLI2 on androgen-independent (AI) prostate cancer cell lines. EXPERIMENTAL DESIGN: A tissue microarray was used to characterize differences in GLI2 expression in benign prostate hyperplasia, prostate cancer treated by neoadjuvant hormonal therapy and AI prostate cancer. The effects of GLI2 ASO on PC-3 cell growth and paclitaxel chemosensitivity were assessed in vitro and in vivo. Oligonucleotide spotted microarray analysis was used to determine alteration in GLI2 coregulated genes after ASO treatment. RESULTS: The expression of GLI2 was significantly higher in prostate cancer than in benign prostate hyperplasia, decreased after androgen ablation in a time-dependent fashion, but became highly expressed again in AI prostate cancer. GLI2 ASO treatment of PC-3 cells reduced GLI2 mRNA and protein levels in a dose-dependent manner. GLI2 knockdown increased PC-3 cell apoptotic rates and significantly decreased cell growth and modulated levels of apoptosis-related genes, such as Bcl2, Bcl-xL, and clusterin. GLI2 knockdown also changed levels of several cell cycle regulators, such as cyclin D1, p27, and PKC-eta. Systematic administration of GLI2 ASO in athymic mice significantly delayed PC-3 tumor progression and enhanced paclitaxel chemosensitivity. CONCLUSIONS: These findings suggest that increased levels of GLI2 correlates with AI progression and that GLI2 may be a therapeutic target in castrate-resistant prostate cancer.


Assuntos
Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Nucleares/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Paclitaxel/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Androgênios/farmacologia , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose , Sequência de Bases , Linhagem Celular Tumoral , Progressão da Doença , Relação Dose-Resposta a Droga , Humanos , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Masculino , Proteínas de Membrana/análise , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Hormônio-Dependentes/genética , Neoplasias Hormônio-Dependentes/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Neoplasias da Próstata/genética , Proteínas de Saccharomyces cerevisiae/análise , Proteína Gli2 com Dedos de Zinco
6.
Int J Cancer ; 122(10): 2368-76, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18240145

RESUMO

Therapeutic resistance is the underlying cause for most cancer deaths and a major problem associated with treatment of metastatic prostate cancer. HTI-286, a fully synthetic analog of the natural tripeptide hemiasterlin, inhibits tubulin polymerization and circumvents transport-based resistance to taxanes. In our study, we evaluated its inhibitory effects on human prostate cancer growth in vitro and in different in vivo models. Androgen-dependent and androgen-independent prostate cancer cell lines including a docetaxel-refractory PC-3 subline (PC-3dR) were treated with HTI-286. Transcriptional profiling was carried out to screen for changes in gene expression induced by HTI-286 and compared to docetaxel. In vivo, nude mice with established PC-3 or PC-3dR xenografts were given HTI-286 intravenously. Additionally, mice bearing hormone-sensitive LNCaP tumors were treated with castration in combination with early or delayed HTI-286 therapy. In all cell lines tested, HTI-286 was a potent inhibitor of proliferation and induced marked increases in apoptosis. Despite similar transcriptomic changes regarding cell death and cell cycle regulating genes after exposure to HTI-286 or docetaxel, array analysis revealed distinct molecular signatures for both compounds. Invivo, HTI-286 significantly inhibited growth of PC-3 and LNCaP xenografts and retained potency in PC-3dR tumors. Simultaneous castration plus HTI-286 therapy was superior to sequential treatment in the LNCaP model. In conclusion, HTI-286 showed strong antitumor activity both in androgen-dependent and androgen- independent tumors and may be a promising agent in second- line treatment strategies for patients suffering from docetaxel- refractory prostate cancer.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Docetaxel , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Radiossensibilizantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxoides/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cancer Med ; 7(7): 3385-3392, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29905005

RESUMO

Development of neuroendocrine prostate cancer (NEPC) is emerging as a major problem in clinical management of advanced prostate cancer (PCa). As increasingly potent androgen receptor (AR)-targeting antiandrogens are more widely used, PCa transdifferentiation into AR-independent NEPC as a mechanism of treatment resistance becomes more common and precarious, since NEPC is a lethal PCa subtype urgently requiring effective therapy. Reprogrammed glucose metabolism of cancers, that is elevated aerobic glycolysis involving increased lactic acid production/secretion, plays a key role in multiple cancer-promoting processes and has been implicated in therapeutics development. Here, we examined NEPC glucose metabolism using our unique panel of patient-derived xenograft PCa models and patient tumors. By calculating metabolic pathway scores using gene expression data, we found that elevated glycolysis coupled to increased lactic acid production/secretion is an important metabolic feature of NEPC. Specific inhibition of expression of MCT4 (a plasma membrane lactic acid transporter) by antisense oligonucleotides led to reduced lactic acid secretion as well as reduced glucose metabolism and NEPC cell proliferation. Taken together, our results indicate that elevated glycolysis coupled to excessive MCT4-mediated lactic acid secretion is clinically relevant and functionally important to NEPC. Inhibition of MCT4 expression appears to be a promising therapeutic strategy for NEPC.

8.
Cancer Res ; 65(23): 11083-93, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16322258

RESUMO

One strategy to improve therapies in prostate cancer involves targeting cytoprotective genes activated by androgen withdrawal to delay the emergence of the androgen-independent (AI) phenotype. The objectives of this study were to define changes in Hsp27 levels after androgen ablation and to evaluate the functional relevance of these changes in AI progression. Using a tissue microarray of 232 specimens of hormone-naïve and post-hormone ablation-treated prostate cancer, we found that Hsp27 levels increase after androgen ablation to become highly expressed (>4-fold, P < or = 0.01) in AI tumors. Hsp27 overexpression rendered LNCaP cells highly resistant to androgen withdrawal both in vitro and in vivo. Tumor volume and serum prostate-specific antigen levels increased 4.3- and 10-fold faster after castration when Hsp27 was overexpressed. Treatment of LNCaP tumor cells in vitro with Hsp27 antisense oligonucleotides (ASO) or short-interfering RNA suppressed Hsp27 levels in a dose-dependent and sequence-specific manner increased the apoptotic sub-G0-G1 fraction and caspase-3 cleavage >2-fold, as well as decreased signal transducers and activators of transcription 3 (Stat3) levels and its downstream genes, c-fos and sPLA-2. The cytoprotection afforded by Hsp27 overexpression was attenuated by Stat3 knockdown using specific Stat3 ASO. Coimmunoprecipitation and immunofluorescence confirmed that Hsp27 interacts with Stat3 and that Stat3 levels correlated directly with Hsp27 levels. Hsp27 ASO treatment in athymic mice bearing LNCaP tumors significantly delayed LNCaP tumor growth after castration, decreasing mean tumor volume and serum prostate-specific antigen levels by 57% and 69%, respectively. These findings identify Hsp27 as a modulator of Stat3-regulated apoptosis after androgen ablation and as a potential therapeutic target in advanced prostate cancer.


Assuntos
Proteínas de Choque Térmico/biossíntese , Proteínas de Neoplasias/biossíntese , Neoplasias da Próstata/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Apoptose/genética , Caspase 3 , Caspases/metabolismo , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Progressão da Doença , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Chaperonas Moleculares , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Neoplasias Hormônio-Dependentes/cirurgia , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , Orquiectomia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Oncotarget ; 8(16): 25928-25941, 2017 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-28460430

RESUMO

To avoid over- or under-treatment of primary prostate tumours, there is a critical need for molecular signatures to discriminate indolent from aggressive, lethal disease. Reprogrammed energy metabolism is an important hallmark of cancer, and abnormal metabolic characteristics of cancers have been implicated as potential diagnostic/prognostic signatures. While genomic and transcriptomic heterogeneity of prostate cancer is well documented and associated with tumour progression, less is known about metabolic heterogeneity of the disease. Using a panel of high fidelity patient-derived xenograft (PDX) models derived from hormone-naïve prostate cancer, we demonstrated heterogeneity of expression of genes involved in cellular energetics and macromolecular biosynthesis. Such heterogeneity was also observed in clinical, treatment-naïve prostate cancers by analyzing the transcriptome sequencing data. Importantly, a metabolic gene signature of increased one-carbon metabolism or decreased proline degradation was identified to be associated with significantly decreased biochemical disease-free patient survival. These results suggest that metabolic heterogeneity of hormone-naïve prostate cancer is of biological and clinical importance and motivate further studies to determine the heterogeneity in metabolic flux in the disease that may lead to identification of new signatures for tumour/patient stratification and the development of new strategies and targets for therapy of prostate cancer.


Assuntos
Metabolismo Energético/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Animais , Linhagem Celular Tumoral , Análise por Conglomerados , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Xenoenxertos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Redes e Vias Metabólicas , Camundongos , Prognóstico , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Transcriptoma
10.
Clin Cancer Res ; 11(13): 4905-11, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16000589

RESUMO

PURPOSE: Androgen ablation is the mainstay of systemic therapy for prostate cancer, with cytotoxic therapies reserved for hormone-refractory disease. It is not clear, however, that this is the most appropriate sequence of interventions for this disease. This study addresses the ideal timing of systemic treatments in the Shionogi and LNCaP xenograft models. We explored the hypothesis that stress-induced gene expression changes after chemotherapy can induce a hormone-independent phenotype. EXPERIMENTAL DESIGN: Three groups of mice bearing either Shionogi or LNCaP xenografts were treated with (a) initial castration and delayed paclitaxel, (b) initial paclitaxel and delayed castration, or (c) simultaneous castration plus paclitaxel. End points were time to tumor progression and time to sacrifice. Microarray and reverse transcription-PCR analyses were carried out to assess changes in gene expression induced by paclitaxel. RESULTS: Mice receiving simultaneous therapy showed a significant improvement in median time to progression (TTP: Shionogi, 65 versus 38 days, P = 0.004; LNCaP, 105 versus 70 days, P = 0.032) and time to sacrifice (Shionogi, 83 versus 66 days, P < 0.014) versus best sequential therapy. A marked lack of response to castration was observed after initial paclitaxel therapy. Gene expression and reverse transcription-PCR studies confirmed that several genes known to play a role in androgen independence were up-regulated in response to paclitaxel exposure. CONCLUSIONS: In laboratory models of prostate cancer, simultaneous androgen deprivation plus paclitaxel is more effective than sequential treatments. These findings provide preclinical proof-of-principle for ongoing clinical trials addressing the role and timing of systemic therapies in prostate cancer.


Assuntos
Orquiectomia , Paclitaxel/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Androgênios/deficiência , Androgênios/farmacologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Terapia Combinada , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Paclitaxel/uso terapêutico , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
11.
J Am Vet Med Assoc ; 228(7): 1053-62, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16579784

RESUMO

OBJECTIVE: To determine whether argyrophilic nucleolar organizing regions (AgNORs), Ki-67, and proliferating cell nuclear antigen (PCNA) scores were associated with histologic grade and survival in dogs with soft tissue sarcomas (STSs). DESIGN: Retrospective study. ANIMALS: 60 dogs with STSs. PROCEDURE: Medical records were examined and histologic specimens were reviewed. Tissue specimens obtained from archival materials were used to prepare sections for histologic staining for AgNOR and immunohistochemical staining for Ki-67 and PCNA labeling. Follow-up monitoring was obtained by reevaluation or telephone conversations with referring veterinarians or owners. RESULTS: 27 (45%) STSs were grade 1, 23 (38%) were grade 2, and 10 (17%) were grade 3. The mean and median AgNOR, Ki-67, and PCNA scores were determined, and significant positive associations among AgNOR and Ki-67 scores with histologic grade and mitotic score were detected. Fifty-four dogs had adequate follow-up examinations and were included in survival analysis and evaluation of prognostic factors. Overall median survival time was > 1,306 days. Twelve of 54 (22%) dogs died of tumor-related causes. Metastatic disease developed in 8 of 54 (15%) dogs. Results of univariate analysis indicated that increased mitotic score, increased AgNOR score, increased Ki-67 score, incomplete surgical margins, noncurative intent surgery, Ki-67 score greater than the median Ki-67 score, and AgNOR score greater than the median AgNOR score were prognostic factors for decreased survival time. Results of multivariate analysis indicated that increased AgNOR score was the only prognostic factor for decreased survival time. CONCLUSIONS AND CLINICAL RELEVANCE: Results suggested that AgNORs and possibly Ki-67 should be routinely evaluated with histologic grading for STSs in dogs.


Assuntos
Antígenos Nucleares/metabolismo , Doenças do Cão/patologia , Antígeno Ki-67/metabolismo , Estadiamento de Neoplasias/veterinária , Proteínas Nucleares/metabolismo , Sarcoma/veterinária , Neoplasias de Tecidos Moles/veterinária , Animais , Antígenos Nucleares/análise , Doenças do Cão/metabolismo , Doenças do Cão/mortalidade , Cães , Feminino , Antígeno Ki-67/análise , Masculino , Análise Multivariada , Proteínas Nucleares/análise , Prognóstico , Antígeno Nuclear de Célula em Proliferação/análise , Antígeno Nuclear de Célula em Proliferação/metabolismo , Estudos Retrospectivos , Sarcoma/metabolismo , Sarcoma/mortalidade , Sarcoma/patologia , Neoplasias de Tecidos Moles/metabolismo , Neoplasias de Tecidos Moles/mortalidade , Neoplasias de Tecidos Moles/patologia , Análise de Sobrevida
12.
Cancer Res ; 64(6): 2212-21, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15026365

RESUMO

Androgen ablation, the most common therapeutic treatment used for advanced prostate cancer, triggers the apoptotic regression of prostate tumors. However, remissions are temporary because surviving prostate cancer cells adapt to the androgen-deprived environment and form androgen-independent (AI) tumors. We hypothesize that adaptive responses of surviving tumor cells result from dysregulated gene expression of key cell survival pathways. Therefore, we examined temporal alterations to gene expression profiles in prostate cancer during progression to androgen independence at several time points using the LNCaP xenograft tumor model. Two key genes, sterol response element-binding protein (SREBP)-1 and -2 (SREBP-1a,-1c, and -2), were consistently dysregulated. These genes are known to coordinately control the expression of the groups of enzymes responsible for lipid and cholesterol synthesis. Northern blots revealed modest increased expression of SREBP-1a, -1c, and -2 after castration, and at androgen independence (day 21-28), the expression levels of both SREBP-1a and -1c were significantly greater than precastrate levels. Changes in SREBP-1 and -2 protein expression were observed by Western analysis. SREBP-1 68-kDa protein levels were maintained throughout progression, however, SREBP-2 68-kDa protein expression increased after castration and during progression (3-fold). SREBPs are transcriptional regulators of over 20 functionally related enzymes that coordinately control the metabolic pathways of lipogenesis and cholesterol synthesis, some of which were likewise dysregulated during progression to androgen independence. RNA levels of acyl-CoA-binding protein/diazepam-binding inhibitor and fatty acid synthase decreased significantly after castration, and then, during progression, increased to levels greater than or equal to precastrate levels. Expression of farnesyl diphosphate synthase did not decrease after castration but did increase significantly during progression to androgen independence. Levels of SREBP cleavage-activating protein, a regulator of SREBP transcriptional activity, decreased after castration and increased significantly at androgen independence. In clinical prostate cancer specimens from patients with varying grades of disease, the stained tissue sections showed high levels of SREBP-1 protein compared with noncancerous prostate tissue. After hormone withdrawal therapy, tumor levels of SREBP-1 decreased significantly after 6 weeks. AI tumors expressed significantly higher levels of SREBP-1. In summary, the LNCaP xenograft model of human prostate cancer as well as clinical specimens of prostate cancer demonstrated an up-regulation of SREBPs and their downstream effector genes during progression to androgen independence. As the AI phenotype emerges, enzymes critical for lipogenesis and cholesterol synthesis are activated and likely contribute significantly to cell survival of AI prostate cancer.


Assuntos
Androgênios/fisiologia , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias da Próstata/metabolismo , Fatores de Transcrição/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteínas Estimuladoras de Ligação a CCAAT/genética , Divisão Celular/efeitos dos fármacos , Colesterol/metabolismo , Proteínas de Ligação a DNA/genética , Progressão da Doença , Humanos , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Terapia Neoadjuvante , Neoplasias Hormônio-Dependentes/patologia , Neoplasias Hormônio-Dependentes/terapia , Orquiectomia , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , RNA Mensageiro/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1 , Proteína de Ligação a Elemento Regulador de Esterol 2 , Esteróis/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos
13.
EMBO Mol Med ; 8(7): 761-78, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27198502

RESUMO

Clusterin (CLU) is a stress-activated molecular chaperone that confers treatment resistance to taxanes when highly expressed. While CLU inhibition potentiates activity of taxanes and other anti-cancer therapies in preclinical models, progression to treatment-resistant disease still occurs implicating additional compensatory survival mechanisms. Taxanes are believed to selectively target cells in mitosis, a complex mechanism controlled in part by balancing antagonistic roles of Cdc25C and Wee1 in mitosis progression. Our data indicate that CLU silencing induces a constitutive activation of Cdc25C, which delays mitotic exit and hence sensitizes cancer cells to mitotic-targeting agents such as taxanes. Unchecked Cdc25C activation leads to mitotic catastrophe and cell death unless cells up-regulate protective mechanisms mediated through the cell cycle regulators Wee1 and Cdk1. In this study, we show that CLU silencing induces a constitutive activation of Cdc25C via the phosphatase PP2A leading to relief of negative feedback inhibition and activation of Wee1-Cdk1 to promote survival and limit therapeutic efficacy. Simultaneous inhibition of CLU-regulated cell cycle effector Wee1 may improve synergistic responses of biologically rational combinatorial regimens using taxanes and CLU inhibitors.


Assuntos
Antineoplásicos/farmacologia , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Proliferação de Células/efeitos dos fármacos , Clusterina/metabolismo , Mitose/efeitos dos fármacos , Neoplasias da Próstata/patologia , Taxoides/farmacologia , Linhagem Celular Tumoral , Clusterina/genética , Técnicas de Silenciamento de Genes , Humanos , Masculino
14.
Neoplasia ; 17(11): 805-16, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26678908

RESUMO

Vascular endothelial growth factor (VEGF)-targeted antiangiogenic therapy significantly inhibits the growth of clear cell renal cell carcinoma (RCC). Eventually, therapy resistance develops in even the most responsive cases, but the mechanisms of resistance remain unclear. Herein, we developed two tumor models derived from an RCC cell line by conditioning the parental cells to two different stresses caused by VEGF-targeted therapy (sunitinib exposure and hypoxia) to investigate the mechanism of resistance to such therapy in RCC. Sunitinib-conditioned Caki-1 cells in vitro did not show resistance to sunitinib compared with parental cells, but when tested in vivo, these cells appeared to be highly resistant to sunitinib treatment. Hypoxia-conditioned Caki-1 cells are more resistant to hypoxia and have increased vascularity due to the upregulation of VEGF production; however, they did not develop sunitinib resistance either in vitro or in vivo. Human endothelial cells were more proliferative and showed increased tube formation in conditioned media from sunitinib-conditioned Caki-1 cells compared with parental cells. Gene expression profiling using RNA microarrays revealed that several genes related to tissue development and remodeling, including the development and migration of endothelial cells, were upregulated in sunitinib-conditioned Caki-1 cells compared with parental and hypoxia-conditioned cells. These findings suggest that evasive resistance to VEGF-targeted therapy is acquired by activation of VEGF-independent angiogenesis pathways induced through interactions with VEGF-targeted drugs, but not by hypoxia. These results emphasize that increased inhibition of tumor angiogenesis is required to delay the development of resistance to antiangiogenic therapy and maintain the therapeutic response in RCC.


Assuntos
Inibidores da Angiogênese/metabolismo , Carcinoma de Células Renais/metabolismo , Sistemas de Liberação de Medicamentos , Indóis/metabolismo , Neoplasias Renais/metabolismo , Pirróis/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/administração & dosagem , Animais , Carcinoma de Células Renais/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos/métodos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/fisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Indóis/administração & dosagem , Neoplasias Renais/tratamento farmacológico , Camundongos , Camundongos Nus , Pirróis/administração & dosagem , Sunitinibe , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
15.
Oncotarget ; 6(33): 34818-30, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26472187

RESUMO

Tumor microenvironments are characterized by decreased oxygen and nutrition due to the rapid and progressive nature of tumors and also stresses induced by several anti-tumor therapies. These intense cell stressors trigger a protective cell survival mechanism heralded by the unfolded protein response (UPR). The UPR is induced by an accumulation of unfolded proteins in the endoplasmic reticulum (ER) following cell starvation. Although the ER stress response is implicated in cytoprotection, its precise role during anti-angiogenic therapy remains unclear. One of the major proteins involved in ER stress is glucose-regulated protein 78 (GRP78), which binds to unfolded proteins and dissociates from membrane-bound ER stress sensors. To determine the role of ER stress responses during anti-angiogenic therapy and the potential role of GRP78 in combined therapy in renal cell carcinoma (RCC), we used GRP78 overexpressing or knockdown RCC cells under hypoxic or hypoglycemic conditions in vitro and in animal models treated with sunitinib. Here, we report that GRP78 plays a crucial role in protecting RCC cells from hypoxic and hypoglycemic stress induced by anti-angiogenic therapy. Knockdown of GRP78 using siRNA inhibited cancer cell survival and induced apoptosis in RCC cells in vitro and also resulted in ER stress-induced apoptosis and hypoxic/hypoglycemic stress-induced apoptosis by inactivating the PERK/eIF-2α pathway. Finally, GRP78 knockdown showed potent suppression of tumor growth and enhanced the antitumor effect of sunitinib in RCC xenografts. Our findings suggest that GRP78 may serve as a novel therapeutic target in combination with anti-angiogenic therapy for the management of RCC.


Assuntos
Carcinoma de Células Renais/patologia , Estresse do Retículo Endoplasmático/fisiologia , Proteínas de Choque Térmico/metabolismo , Neoplasias Renais/patologia , eIF-2 Quinase/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Western Blotting , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Imunofluorescência , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Indóis/farmacologia , Camundongos , Camundongos Nus , Microscopia Confocal , Pirróis/farmacologia , RNA Interferente Pequeno , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Estresse Fisiológico , Sunitinibe , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Mol Cancer Ther ; 14(4): 1024-34, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25657336

RESUMO

Renal cell carcinoma (RCC) is the most common malignancy in the kidney. Antiangiogenic targeted therapies inhibit the progression of RCC, but have limited impacts on invasion or metastasis of tumor cells. Integrin-linked kinase (ILK) is a serine/threonine kinase implicated in the regulation of cell growth/survival, cell-cycle progression, epithelial-mesenchymal transition (EMT), invasion/migration, and angiogenesis. However, the role of ILK in RCC has not been evaluated. We investigated the role of ILK on cancer progression and metastasis and the therapeutic potential of ILK inhibition in RCC. Our investigation reveals that ILK is expressed at a low level in normal cells and low-stage RCC cells and is highly expressed in advanced and metastatic cells. Caki-1, a metastatic RCC cell line, showed higher expression of molecular EMT markers, including Snail and Zeb1, but decreased activity of GSK3ß. Knockdown of ILK using small interference (si)-ILK minimally inhibited tumor proliferation and cell-cycle progression was not significantly affected. However, ILK knockdown suppressed the formation of stress fibers and focal adhesions and impeded phenotypic EMT markers, including cell migration and invasion, in Caki-1 and UMRC-3 cells. Finally, in vivo knockdown of ILK suppressed the progression, invasion, and metastasis of primary RCC in nude mice by downregulation of EMT markers (Snail, Zeb1, vimentin, and E-cadherin). Our results show that ILK may be essential for invasion and metastasis in RCC and regulates vimentin and E-cadherin expression by regulating the EMT-related transcription factors Snail and Zeb1. These results suggest that ILK may be a potential target in RCC.


Assuntos
Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Transição Epitelial-Mesenquimal/genética , Proteínas Serina-Treonina Quinases/genética , Animais , Biomarcadores , Carcinoma de Células Renais/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Sobrevivência Celular/genética , Modelos Animais de Doenças , Regulação para Baixo , Expressão Gênica , Técnicas de Silenciamento de Genes , Xenoenxertos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Estadiamento de Neoplasias , Fenótipo , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco
17.
Clin Tech Small Anim Pract ; 18(2): 92-7, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12831068

RESUMO

Canine lymphoma is one of the most commonly diagnosed canine neoplasms. It is helpful to classify lymphoma anatomically, because these forms each have common histories and clinical signs. Anatomic forms include multicentric, alimentary, mediastinal, and cutaneous forms. Because lymphoma is a systemic disease, systemic chemotherapy is the most appropriate modality for its treatment. Lymphoma cells are sensitive to chemotherapy, and complete remission rates are high when these patients are treated with conventional chemotherapy. Treated dogs maintain a good quality of life, and treatment can provide resolution of many presenting signs and abnormalities. The fundamental goals of chemotherapy are to induce a durable remission and to re-induce a remission after one or more relapses. Other therapies, such as surgery and radiation therapy, are appropriate in certain situations. Prognostic factors will also be summarized.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Doenças do Cão/tratamento farmacológico , Linfoma/veterinária , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Terapia Combinada/veterinária , Doenças do Cão/patologia , Doenças do Cão/radioterapia , Doenças do Cão/cirurgia , Cães , Linfoma/tratamento farmacológico , Estadiamento de Neoplasias/veterinária , Prognóstico , Medicina Veterinária
18.
Clin Tech Small Anim Pract ; 18(2): 98-102, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12831069

RESUMO

Lymphoma is the most commonly diagnosed neoplasm in cats. As feline leukemia virus antigenemia has decreased over the past 15 years, there has been a profound shift in the presence, signalment, and frequency of sites of feline lymphoma in North America. There is variation in anatomic classification systems, but most studies have divided lymphoma into four groups: alimentary, mediastinal, multicentric, or extranodal. Clinical signs and common differential diagnoses for each of the forms are described. Staging allows for evaluation of the extent of disease. As in the dog, lymphoma is a systemic disease in the cat, and chemotherapy is the treatment of choice for most forms. Exceptions are described. In contrast to canine lymphoma, feline lymphoma is generally more challenging and frustrating to treat than canine lymphoma. Response rates are lower, and remission duration is shorter. Fortunately, cats treated with chemotherapy tend to have less toxicity than dogs. Positive prognostic factors are feline leukemia virus-negative, clinically well at time of diagnosis, and response to therapy. Achieving a complete remission is prognostic for survival. Unfortunately, response cannot be predicted before treatment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Doenças do Gato/tratamento farmacológico , Linfoma/veterinária , Animais , Doenças do Gato/patologia , Doenças do Gato/radioterapia , Doenças do Gato/cirurgia , Gatos , Terapia Combinada/veterinária , Linfoma/tratamento farmacológico , Estadiamento de Neoplasias/veterinária , Medicina Veterinária
19.
Clin Tech Small Anim Pract ; 18(2): 118-22, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12831074

RESUMO

Soft-tissue sarcomas develop from a variety of mesenchymal tissues, but they are often considered collectively, due to similarity in clinical behavior and histologic features. These tumors are locally invasive, with poorly defined histologic margins and neoplastic cells that often infiltrate through fascial planes. In general, local recurrence is common following conservative excision. Pretreatment biopsy provides information on tumor type and grade, which will allow the clinician to properly plan for an aggressive first surgery. Adopted from human medicine, the canine histopathologic grading system is predictive. Specifically, mitotic rate is predictive for metastasis, and necrosis and mitotic rate are predictive for survival. Diagnostic imaging is useful to determine the extent of disease and for treatment planning. The most effective treatment for soft-tissue sarcomas is surgical excision. Surgery with curative intent requires preoperative biopsy, planning, and a wide first excision. Increasingly, surgery is being replaced by a combined-modality approach. Radiation therapy plays an important role in the management of soft-tissue sarcomas, but it has little role as a single treatment modality. Radiation therapy is appropriate for incompletely excised tumors or for preoperative treatment. Chemotherapy's role is most appropriate in the adjunct setting, and is mainly used to treat incompletely resected tumors, high-grade tumors, and metastatic disease.


Assuntos
Doenças do Cão/terapia , Sarcoma/veterinária , Neoplasias de Tecidos Moles/veterinária , Animais , Terapia Combinada/veterinária , Doenças do Cão/diagnóstico , Doenças do Cão/tratamento farmacológico , Doenças do Cão/radioterapia , Doenças do Cão/cirurgia , Cães , Prognóstico , Sarcoma/terapia , Neoplasias de Tecidos Moles/terapia , Medicina Veterinária
20.
Endocr Relat Cancer ; 20(2): 173-86, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23319492

RESUMO

IGF2 is a mitogenic foetal growth factor commonly over-expressed in cancers, including prostate cancer (PC). We recently demonstrated that insulin can activate de novo steroidogenesis in PC cells, a major pathway for reactivation of androgen pathways and PC progression. IGF2 can activate the IGF1 receptor (IGF1R) or insulin receptor (INSR) or hybrids of these two receptors. We therefore hypothesized that IGF2 may contribute to PC progression via de novo steroidogenesis. IGF2 mRNA but not IGF2 receptor mRNA expression was increased in patient samples during progression to castrate-resistant PC as was immunoreactivity to INSR and IGF1R antibodies. Treatment of androgen receptor (AR)-positive PC cell lines LNCaP and 22RV1 with IGF2 for 48 h resulted in increased expression of steroidogenic enzyme mRNA and protein, including steroid acute regulatory protein (StAR), cytochrome p450 family member (CYP)17A1, aldo-keto reductase family member (AKR)1C3 and hydroxysteroid dehydrogenase (HSD)17B3. IGF2 treatment resulted in increased steady state steroid levels and increased de novo steroidogenesis resulting in AR activation as demonstrated by PSA mRNA induction. Inhibition of the IGF1R/INSR signalling axis attenuated the effects of IGF2 on steroid hormone synthesis. We present a potential mechanism for prostatic IGF2 contributing to PC progression by inducing steroidogenesis and that IGF2 signalling and related pathways present attractive targets for PC therapy.


Assuntos
Fator de Crescimento Insulin-Like II/farmacologia , Neoplasias da Próstata/metabolismo , Esteroides/biossíntese , Linhagem Celular Tumoral , Humanos , Fator de Crescimento Insulin-Like II/genética , Masculino , Antígeno Prostático Específico/genética , RNA Mensageiro/metabolismo , Receptor IGF Tipo 2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA