Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Br J Pharmacol ; 177(2): 328-345, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31621898

RESUMO

BACKGROUND AND PURPOSE: Fenretinide, a synthetic retinoid derivative first investigated for cancer prevention and treatment, has been shown to ameliorate glucose tolerance, improve plasma lipid profile and reduce body fat mass. These effects, together with its ability to inhibit ceramide synthesis, suggest that fenretinide may have an anti-atherosclerotic action. EXPERIMENTAL APPROACH: To this aim, nine-week-old apoE-knockout (EKO) female mice were fed for twelve weeks a Western diet, without (control) or with (0.1% w/w) fenretinide. As a reference, wild-type (WT) mice were treated similarly. Growth and metabolic parameters were monitored throughout the study. Atherosclerosis development was evaluated in the aorta and at the aortic sinus. Blood and lymphoid organs were further characterized with thorough cytological/histological and immunocytofluorimetric analyses. KEY RESULTS: Fenretinide treatment significantly lowered body weight, glucose levels and plasma levels of total cholesterol, triglycerides, and phospholipids. In the liver, fenretinide remarkably reduced hepatic glycogenosis and steatosis driven by the Western diet. Treated spleens were abnormally enlarged, with severe follicular atrophy and massive extramedullary haematopoiesis. Severe renal hemosiderin deposition was observed in treated EKO mice. Treatment resulted in a threefold increase of total leukocytes (WT and EKO) and raised the activated/resting monocyte ratio in EKO mice. Finally, atherosclerosis development was markedly increased at the aortic arch, thoracic and abdominal aorta of fenretinide-treated mice. CONCLUSIONS AND IMPLICATIONS: We provide the first evidence that, despite beneficial metabolic effects, fenretinide treatment may enhance the development of atherosclerosis.


Assuntos
Antineoplásicos/toxicidade , Aorta/efeitos dos fármacos , Doenças da Aorta/induzido quimicamente , Aterosclerose/induzido quimicamente , Metabolismo Energético/efeitos dos fármacos , Fenretinida/toxicidade , Animais , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Dieta Ocidental , Modelos Animais de Doenças , Progressão da Doença , Feminino , Lipídeos/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Placa Aterosclerótica , Baço/efeitos dos fármacos , Baço/metabolismo , Baço/patologia , Redução de Peso/efeitos dos fármacos
2.
Neurotoxicology ; 73: 258-264, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30980846

RESUMO

Traditional approaches (e.g., neurobehavior, neuropathology) can detect alterations in apical endpoints indicative of developmental neurotoxicity (DNT). However, there is an increasing desire to understand mode-of-action (MOA) for DNT effects; thus, this short communication describes initial work on a neuronal differentiation assay. Basically, our laboratory used the human NT2/D1 cell line to develop an assay to evaluate toxicants for effects on all-trans retinoic acid (RA)-induced neuronal differentiation. Based on literature reports, we selected a neuronal protein, neuronal class III ß-tubulin (ß3-tubulin), as a marker of differentiation. For this assay, cultured RA-treated NT2 cells were trypsinized to individual cells, methanol fixed, and labeled with a ß3-tubulin specific monoclonal antibody (TUJ1). Characterization studies using 100,000 cells/sample showed that NT2 cells had appreciable expression of ß3-tubulin starting around day 7 of the differentiation process with a peak expression noted around day 12. Methylmercury, 22(R)-hydroxycholesterol, N-(4-hydroxyphenol)retinamide (4HPR), and 9-cis retinoic acid were selected as initial test compounds. Of these, only 9-cis RA, which is known to affect the RA pathway, was positive for specific impacts on differentiation. These results demonstrate the feasibility of using a flow cytometry method targeting specific cellular biomarkers for evaluating effects on neuronal differentiation. Additional assays are needed to detect compounds targeting other (non-RA) neuronal differentiation pathways. Ultimately, a battery of in vitro assays would be needed to evaluate the potential MOAs involved in altered neuronal differentiation.


Assuntos
Alitretinoína/toxicidade , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Testes de Toxicidade , Tretinoína/farmacologia , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fenretinida/toxicidade , Citometria de Fluxo , Humanos , Hidroxicolesteróis/toxicidade , Compostos de Metilmercúrio/toxicidade , Neurônios/metabolismo , Neurônios/patologia , Medição de Risco , Transdução de Sinais , Fatores de Tempo , Tubulina (Proteína)/metabolismo
3.
Toxicol Sci ; 161(1): 58-75, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28973697

RESUMO

Pharmaceuticals and chemicals produce hemangiosarcomas (HS) in mice, often by nongenotoxic, proliferative mechanisms. A mode-of-action (MOA) for hemangiosarcoma was proposed based on information presented at an international workshop (Cohen et al., Hemangiosarcoma in rodents: Mode-of-action evaluation and human relevance. Toxicol. Sci. 111, 4-18.). Five key elements of the MOA were articulated and included hypoxia, macrophage activation, increased angiogenic growth factors, dysregulated angiogenesis/erythropoiesis, and endothial cell proliferation. The goal of the current study was to add to the weight-of-evidence for the proposed MOA by assessing these key elements with 3 different compounds of varying potency for HS induction: fenretinide (high), troglitazone (intermediate), and elmiron (low). Multiple endpoints, including hypoxia (hyproxyprobe, transcriptomics), endothelial cell (EC) proliferation, and clinical and anatomic pathology, were assessed after 2, 4, and 13-weeks of treatment in B6C3F1 mice. All 3 compounds demonstrated strong evidence for dysregulated erythropoiesis (decrease in RBC and a failure to increase reticulocytes) and macrophage activation (4- to 11-fold increases); this pattern of hematological changes in mice might serve as an early biomarker to evaluate EC proliferation in suspected target organs for potential HS formation. Fenretinide demonstrated all 5 key elements, while troglitazone demonstrated 4 and elmiron demonstrated 3. Transcriptomics provided support for the 5 elements of the MOA, but was not any more sensitive than hypoxyprobe immunohistochemistry for detecting hypoxia. The overall transcriptional evidence for the key elements of the proposed MOA was also consistent with the potency of HS induction. These data, coupled with the previous work with 2-butoxyethanol and pregablin, increase the weight-of-evidence for the proposed MOA for HS formation.


Assuntos
Fenretinida/toxicidade , Hemangiossarcoma/induzido quimicamente , Neovascularização Patológica/induzido quimicamente , Poliéster Sulfúrico de Pentosana/toxicidade , Troglitazona/toxicidade , Animais , Hipóxia Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Hemangiossarcoma/metabolismo , Hemangiossarcoma/patologia , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Especificidade de Órgãos
4.
Cancer Chemother Pharmacol ; 71(3): 777-87, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23314735

RESUMO

OBJECTIVE: Neuroblastoma is a common, frequently fatal, neural crest tumor of childhood. Chemotherapy-resistant neuroblastoma cells typically have Schwann cell-like ("S-type") morphology and express the p75 neurotrophin receptor (p75NTR). p75NTR has been previously shown to modulate the redox state of neural crest tumor cells. We, therefore, hypothesized that p75NTR expression level would influence the effects of the redox-active chemotherapeutic drug fenretinide on neuroblastoma cells. METHODS: Transfection and lentiviral transduction were used to manipulate p75NTR expression in these cell lines. Sensitivity to fenretinide was determined by concentration- and time-cell survival studies. Apoptosis incidence was determined by morphological assessment and examination of cleavage of poly-ADP ribose polymerase and caspase-3. Generation and subcellular localization of reactive oxygen species were quantified using species- and site-specific stains and by examining the effects of site-selective antioxidants on cell survival after fenretinide treatment. Studies of mitochondrial electron transport employed specific inhibitors of individual proteins in the electron transport chain. RESULTS: Knockdown of p75NTR attenuates fenretinide-induced accumulation of mitochondrial superoxide and apoptosis. Overexpression of p75NTR has the opposite effects. Pretreatment of cells with 2-thenoyltrifluoroacetone or dehydroascorbic acid uniquely prevents mitochondrial superoxide accumulation and cell death after fenretinide treatment, indicating that mitochondrial complex II is the likely site of fenretinide-induced superoxide generation and p75NTR-induced potentiation of these phenomena. CONCLUSION: Modification of expression of p75NTR in a particular neuroblastoma cell line modifies its susceptibility to fenretinide. Enhancers of p75NTR expression or signaling could be potential drugs for use as adjuncts to chemotherapy of neural tumors.


Assuntos
Antineoplásicos/toxicidade , Neoplasias Encefálicas/tratamento farmacológico , Fenretinida/toxicidade , Proteínas do Tecido Nervoso/uso terapêutico , Neuroblastoma/tratamento farmacológico , Receptores de Fator de Crescimento Neural/uso terapêutico , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Transporte de Elétrons/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Humanos , Indicadores e Reagentes , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Neuroblastoma/genética , Oxirredução , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio , Receptores de Fator de Crescimento Neural/fisiologia , Transdução de Sinais/efeitos dos fármacos
5.
PLoS One ; 8(1): e53927, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23326540

RESUMO

Sphingolipids play a role in the development of emphysema and ceramide levels are increased in experimental models of emphysema; however, the mechanisms of ceramide-related pulmonary emphysema are not fully understood. Here we examine mechanisms of ceramide-induced pulmonary emphysema. Male Sprague-Dawley rats were treated with fenretinide (20 mg/kg BW), a synthetic derivative of retinoic acid that causes the formation of ceramide, and we postulated that the effects of fenretinide could be offset by administering sphingosine 1-phosphate (S1P) (100 µg/kg BW). Lung tissues were analyzed and mean alveolar airspace area, total length of the alveolar perimeter and the number of caspase-3 positive cells were measured. Hypoxia-inducible factor alpha (HIF-1α), vascular endothelial growth factor (VEGF) and other related proteins were analyzed by Western blot analysis. Immunohistochemical analysis of HIF-1α was also performed. Ceramide, dihydroceramide, S1P, and dihydro-S1P were measured by mass spectrometer. Chronic intraperitoneal injection of fenretinide increased the alveolar airspace surface area and increased the number of caspase-3 positive cells in rat lungs. Fenretinide also suppressed HIF-1α and VEGF protein expression in rat lungs. Concomitant injection of S1P prevented the decrease in the expression of HIF-1α, VEGF, histone deacetylase 2 (HDAC2), and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) protein expression in the lungs. S1P injection also increased phosphorylated sphingosine kinase 1. Dihydroceramide was significantly increased by fenretinide injection and S1P treatment prevented the increase in dihydroceramide levels in rat lungs. These data support the concept that increased de novo ceramide production causes alveolar septal cell apoptosis and causes emphysema via suppressing HIF-1α. Concomitant treatment with S1P normalizes the ceramide-S1P balance in the rat lungs and increases HIF-1α protein expression via activation of sphingosine kinase 1; as a consequence, S1P salvages fenretinide induced emphysema in rat lungs.


Assuntos
Ceramidas/biossíntese , Enfisema/tratamento farmacológico , Enfisema/metabolismo , Lisofosfolipídeos/administração & dosagem , Alvéolos Pulmonares/metabolismo , Esfingosina/análogos & derivados , Animais , Caspase 3/metabolismo , Ceramidas/metabolismo , Enfisema/induzido quimicamente , Fenretinida/química , Fenretinida/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Histona Desacetilase 2/metabolismo , Humanos , Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Substâncias Protetoras/administração & dosagem , Alvéolos Pulmonares/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Esfingosina/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Exp Cell Res ; 314(8): 1667-82, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18342855

RESUMO

N-(4-hydroxyphenyl)retinamide (4HPR) is a synthetic retinoid that has been tested in clinical trials as a cancer chemopreventive drug. 4HPR is cytotoxic to cancer cells but the underlying molecular mechanisms are at present only partially understood. Here we demonstrate that in the human cervical cancer cell line HeLa and the human leukemia cell line HL-60, 4HPR caused rapid, Reactive Oxygen Species (ROS)-dependent activation of the Unfolded Protein Response (UPR). In HeLa cells, 4HPR was shown to induce cell death and activation of procaspases. These effects of 4HPR could be abolished by the over-expression of dominant negative mutants of PERK or eIF2 alpha. HeLa cells incubated with 4HPR were found to form autophagosomes that were also mediated by the PERK/eIF2 alpha pathway. While 4HPR-induced cell death could be significantly prevented by the presence of specific caspase inhibitors, 3-methyladenine (3-MA) that inhibits autophagosome formation enhanced 4HPR-induced cell death. Examination of individual 4HPR-treated HeLa cells revealed that those without the development of autophagosomes hence exhibiting an incomplete UPR were caspase-active and were not viable, while those with autophagosomes were caspase-inactive and retained cell viability. Our data suggest that the PERK/eIF2 alpha pathway is essential for the cytotoxicity of 4HPR that targets on cancer cells with malfunctional UPR.


Assuntos
Anticarcinógenos/toxicidade , Fator de Iniciação 2 em Eucariotos/metabolismo , Fenretinida/toxicidade , Transdução de Sinais , eIF-2 Quinase/metabolismo , Fator 6 Ativador da Transcrição/metabolismo , Anticarcinógenos/antagonistas & inibidores , Apoptose , Autofagia , Caspase 3/metabolismo , Caspase 9/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Retículo Endoplasmático/metabolismo , Precursores Enzimáticos/metabolismo , Fator de Iniciação 2 em Eucariotos/genética , Fenretinida/antagonistas & inibidores , Células HL-60 , Células HeLa , Humanos , Chaperonas Moleculares/metabolismo , Mutação , Fagossomos/metabolismo , Dobramento de Proteína , Splicing de RNA , Fatores de Transcrição de Fator Regulador X , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , eIF-2 Quinase/genética
8.
Cell Death Differ ; 11(5): 527-41, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14765134

RESUMO

The majority of ovarian cancer cells are resistant to apoptosis induced by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Subtoxic concentrations of the semisynthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) enhanced TRAIL-mediated apoptosis in ovarian cancer cell lines but not in immortalized nontumorigenic ovarian epithelial cells. The enhancement of TRAIL-mediated apoptosis by 4HPR was not due to changes in the levels of proteins known to modulate TRAIL sensitivity. The combination of 4HPR and TRAIL enhanced cleavage of multiple caspases in the death receptor pathway (including the two initiator caspases, caspase-8 and caspase-9). The 4HPR and TRAIL combination leads to mitochondrial permeability transition, significant increase in cytochrome c release, and increased caspase-9 activation. Caspase-9 may further activate caspase-8, generating an amplification loop. Stable overexpression of Bcl-xL abrogates the interaction between 4HPR and TRAIL at the mitochondrial level by blocking cytochrome c release. As a consequence, a decrease in activation of caspase-9, caspase-8, and TRAIL-mediated apoptosis occurs. These results indicate that the enhancement in TRAIL-mediated apoptosis induced by 4HPR is due to the increase in activation of multiple caspases involving an amplification loop via the mitochondrial-death pathway. These findings offer a promising and novel strategy for the treatment of ovarian cancer.


Assuntos
Apoptose/efeitos dos fármacos , Fenretinida/toxicidade , Glicoproteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Receptores X de Retinoides/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose , Caspases/metabolismo , Citocromos c/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Feminino , Humanos , Ovário/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas
9.
Cell Death Differ ; 11(3): 270-9, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14647238

RESUMO

Fenretinide (HPR), a synthetic retinoid that exhibits lower toxicity than other retinoids, has shown preventive and therapeutic activity against ovarian tumors. Although the growth inhibitory effects of HPR have been ascribed to its ability to induce apoptosis, little is known about the molecular mechanisms involved. Since the proto-oncogene c-Fos has been implicated in apoptosis induction, we analyzed its role in mediating HPR response in a human ovarian carcinoma cell line (A2780) sensitive to HPR apoptotic effect. In these cells, HPR treatment caused induction of c-Fos expression, whereas such an effect was not observed in cells made resistant to HPR-induced apoptosis (A2780/HPR). Moreover, in a panel of other human ovarian carcinoma cell lines, c-Fos inducibility and HPR sensitivity were closely associated. Ceramide, which is involved in HPR-induced apoptosis, was also involved in c-Fos induction because its upregulation by HPR was reduced by fumonisin B(1), a ceramide synthase inhibitor. The causal relationship between c-Fos induction and apoptosis was established by the finding of an increased apoptotic rate in cells overexpressing c-Fos. Similarly to that observed for c-Fos expression, HPR treatment increased c-Jun expression in HPR-sensitive but not in HPR-resistant cells, suggesting the involvement of the transcription factor activating protein 1 (AP-1) in HPR-induced apoptosis. In gene reporter experiments, HPR stimulated AP-1 transcriptional activity and potentiated the AP-1 activity induced by 12-tetradecanoylphorbol 13-acetate. Furthermore, inhibition of AP-1 DNA binding, by transfecting A2780 cells with a dominant-negative Fos gene, caused decreased sensitivity to HPR apoptotic effects. Overall, the results indicate that c-Fos plays a role in mediating HPR-induced growth inhibition and apoptosis in ovarian cancer cells and suggest that c-Fos regulates these processes as a member of the AP-1 transcription factor.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma/tratamento farmacológico , Fenretinida/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Proteínas Proto-Oncogênicas c-fos/metabolismo , Antineoplásicos/toxicidade , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patologia , Linhagem Celular Tumoral , Ceramidas/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Fenretinida/toxicidade , Fumonisinas/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Genes jun/efeitos dos fármacos , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Proto-Oncogene Mas , Fator de Transcrição AP-1/metabolismo , Regulação para Cima
10.
Urology ; 61(5): 1047-52, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12736045

RESUMO

OBJECTIVES: To investigate the feasibility of targeting ceramide metabolism to enhance chemotherapy cytotoxicity in prostate cancer. Discovering new targets for cancer treatment is an important endeavor, especially in prostate malignancies, which often revert to hormone- and chemotherapy-refractory disease states. METHODS: Ceramide metabolism was measured in human prostate cancer cell lines using [(3)H]palmitic acid as the tracer. Cellular lipids were analyzed by thin-layer chromatography and liquid scintillation counting. Cell viability in response to drug exposure was measured spectrophotometrically using commercial cell proliferation reagents. RESULTS: LNCaP cells were five times more sensitive to N-(4-hydroxyphenyl)retinamide (4-HPR), a synthetic retinoid, compared with PC-3 cells. Ceramide levels increased only twofold in PC-3 cells versus 10-fold in LNCaP cells in response to 10 microM 4-HPR. PC-3 resistance to 4-HPR could be reversed by the addition of tamoxifen or other agents that block the metabolism of ceramide to glucosylceramide, and with tamoxifen this was marked by a ninefold increase in cellular ceramide levels. The influence of 4-HPR on ceramide metabolism was shown to be through activation of serine palmitoyltransferase, the rate-limiting enzyme in the ceramide synthesis pathway. Blocking the ceramide generated by 4-HPR reduced the extent of apoptosis. CONCLUSIONS: Increasing intracellular concentrations of ceramide may be an avenue to enhance the cytotoxic response to chemotherapy in human prostate cancer.


Assuntos
Antineoplásicos/farmacologia , Ceramidas/metabolismo , Líquido Intracelular/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Aciltransferases/metabolismo , Antineoplásicos/toxicidade , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ceramidas/biossíntese , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Fenretinida/farmacologia , Fenretinida/toxicidade , Glucosilceramidas/metabolismo , Humanos , Líquido Intracelular/enzimologia , Masculino , Oxirredutases/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/enzimologia , Serina C-Palmitoiltransferase , Tamoxifeno/farmacologia , Tamoxifeno/toxicidade , Células Tumorais Cultivadas
11.
Cancer Invest ; 19(2): 145-54, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11296619

RESUMO

The retinoid 4-(N-hydroxyphenyl)retinamide (4HPR, fenretinide) has both growth inhibitory and apoptosis-inducing effects on a number of cancer cell lines in vitro and in vivo and has been entered into a number of oncological trials. However, little is known about its mechanism(s) of action or its effects on normal cells such as fibroblasts. In this study, the effects of fenretinide on both epidermoid carcinoma cells of vulva (cell line A431) and normal human dermal fibroblasts, both as monolayers and also grown in 3D cell culture systems, have been investigated. The 3D cell culture system contained normal human fibroblasts embedded in a type I collagen gel with the carcinoma cells seeded on top of the collagen gel, which mimics the epidermoid carcinoma. Fenretinide significantly inhibited the rate of DNA synthesis of carcinoma cells, while there was little effect on fibroblasts on monolayers, at 10(-6)-10(-5) M, which are clinically attainable doses. Fenretinide at 5 x 10(-6) M induced apoptosis characterised by cell shrinkage, membrane blebbing, nuclear condensation and/or fragmentation, and cell detachment in carcinoma cells, but not fibroblasts from monolayers. Fenretinide also reduced the viability of carcinoma cells in the 3D cell culture system without affecting fibroblasts. These data show that fenretinide may preferentially induce apoptosis in epidermoid carcinoma cells.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/patologia , Sobrevivência Celular/efeitos dos fármacos , Fenretinida/toxicidade , Pele/efeitos dos fármacos , Animais , Antineoplásicos/toxicidade , Apoptose/fisiologia , Carcinoma de Células Escamosas/ultraestrutura , Técnicas de Cultura de Células/métodos , Colágeno , Fragmentação do DNA , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Nucleossomos/efeitos dos fármacos , Nucleossomos/ultraestrutura , Ratos , Valores de Referência , Pele/citologia , Células Tumorais Cultivadas
12.
Prostate ; 24(6): 299-305, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8208624

RESUMO

The synthetic retinoid N-(4-hydroxyphenyl) retinamide (4-HPR) has been demonstrated to inhibit the development of primary and metastatic neoplasms in several animal models. In order to investigate the effect of 4-HPR on human prostate adenocarcinoma, we designed a series of in vitro experiments with the PC3 cell line to evaluate effects on proliferation, cell cycle kinetics, and c-myc mRNA expression. 4-HPR demonstrated cytotoxicity only at the supraphysiologic concentration of 10.0 microM. However, asynchronously growing cells exposed to 1 microM 4-HPR demonstrated a 51% reduction in proliferation rate, associated with an accumulation of cells in the G0/G1 phase of the cell cycle. PC3 cells synchronized with serum deprivation or aphidicolin exhibited significant decreases in DNA synthesis when treated with 1 microM 4-HPR. Additionally, these cells were found to accumulate in G0/G1 and S phase. Northern blots indicated a significant decrease in c-myc mRNA expression in asynchronously growing cells with continuous administration of 1 microM 4-HPR for 6 days. These data suggest that 4-HPR can inhibit growth of PC3 cells as a consequence of a block in cell cycle transition from G1 to S phase at a concentration of 1 microM, and that this inhibition is associated with a suppression of c-myc gene expression.


Assuntos
Antineoplásicos/toxicidade , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Fenretinida/toxicidade , Adenocarcinoma , Northern Blotting , Linhagem Celular , DNA de Neoplasias/biossíntese , DNA de Neoplasias/efeitos dos fármacos , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Genes myc , Humanos , Masculino , Neoplasias da Próstata , RNA Mensageiro/biossíntese , Timidina/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA