Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Am J Respir Cell Mol Biol ; 51(6): 783-92, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24885263

RESUMEN

Arachidonic acid (AA) and docosahexaenoic acid (DHA) play important roles in inflammation and disease progression, where AA is viewed as proinflammatory and DHA as antiinflammatory. We observe in our model of allergic asthma that the AA/DHA ratio is significantly skewed in a proinflammatory direction. Fenretinide, a vitamin A derivative, has been shown to correct fatty acid imbalances in other diseases. Therefore, we explored if fenretinide can have a protective effect in allergic asthma. To accomplish this, we measured the levels of AA and DHA in the lungs of nonallergic, ovalbumin-induced allergic, and fenretinide-treated allergic mice. We also investigated the effect of allergic asthma and fenretinide treatment on markers of oxidative stress, levels of metabolites, IgE production, airway hyperresponsiveness, and histological changes. Our data demonstrate that treatment of allergen-sensitized mice with fenretinide before allergen challenge prevents ovalbumin-induced changes in the AA/DHA ratio. The levels of several metabolites, such as serotonin, and markers of cellular stress, which are increased after ovalbumin challenge, are also controlled by fenretinide treatment. We observed the protective effect of fenretinide against ovalbumin-induced airway hyperresponsiveness and inflammation in the lungs, illustrated by a complete block in the infiltration of inflammatory cells to the airways and dramatically diminished goblet cell proliferation, even though IgE remained high. Our results demonstrate that fenretinide is an effective agent targeting inflammation, oxidation, and lung pathology observed in allergic asthma.


Asunto(s)
Antiinflamatorios/farmacología , Asma/tratamiento farmacológico , Fenretinida/farmacología , Alérgenos/inmunología , Animales , Ácido Araquidónico/metabolismo , Asma/inmunología , Línea Celular , Ácidos Docosahexaenoicos/metabolismo , Evaluación Preclínica de Medicamentos , Inmunoglobulina E/sangre , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ovalbúmina/inmunología
2.
Infect Immun ; 82(5): 1778-85, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24549326

RESUMEN

Streptococcus suis is an important swine pathogen and an emergent zoonotic pathogen. Excessive inflammation caused by S. suis is responsible for early high mortality in septic shock-like syndrome cases. Polyunsaturated fatty acids (PUFAs) may contribute to regulating inflammatory processes. This study shows that mouse infection by S. suis is accompanied by an increase of arachidonic acid, a proinflammatory omega-6 (ω-6) PUFA, and by a decrease of docosahexaenoic acid, an anti-inflammatory ω-3 PUFA. Macrophages infected with S. suis showed activation of mitogen-activated protein kinase pathways and cyclooxygenase-2 upregulation. Fenretinide, a synthetic vitamin A analog, reduced in vitro expression of inflammatory mediators. Pretreatment of mice with fenretinide significantly improved their survival by reducing systemic proinflammatory cytokines during the acute phase of an S. suis infection. These findings indicate a beneficial effect of fenretinide in diminishing the expression of inflammation and improving survival during an acute infection by a virulent S. suis strain.


Asunto(s)
Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-6/metabolismo , Infecciones Estreptocócicas/metabolismo , Streptococcus suis/fisiología , Animales , Anticarcinógenos/farmacología , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Ácidos Grasos Omega-3/sangre , Ácidos Grasos Omega-6/sangre , Fenretinida/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Infecciones Estreptocócicas/sangre , Zoonosis
3.
Anticancer Drugs ; 24(6): 577-86, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23542749

RESUMEN

The outcome in patients with breast cancer is not satisfactory to date, although new chemotherapy regimens have been introduced in clinics. Therefore, novel approaches are required for better management of patients with breast cancer. In this study, we tested the cytotoxic activity of a new combination of fenretinide, a synthetic retinoid, with indole-3-carbinol, a natural product present in vegetables such as broccoli and cabbage, against MCF-7 (estrogen receptor-positive) and MDA-MB-231 (estrogen receptor-negative) cell lines. It has been found that the combination resulted in more powerful cytotoxic activity, by induction of apoptosis, compared with that when they were used singly. In conclusion, this novel combination warrants in-vivo experiments to elucidate its possible use in the treatment of breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Fenretinida/farmacología , Indoles/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , ADN Complementario/metabolismo , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , ARN/metabolismo , ARN Interferente Pequeño/metabolismo , Transfección
4.
Diabetes ; 62(3): 825-36, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23193184

RESUMEN

The synthetic retinoid, Fenretinide (FEN), inhibits obesity and insulin resistance in mice and is in early clinical trials for treatment of insulin resistance in obese humans. We aimed to determine whether alterations in retinoic acid (RA)-responsive genes contribute to the beneficial effects of FEN. We examined the effect of FEN on 3T3-L1 adipocyte differentiation and alterations in gene expression in C57Bl/6 and retinaldehyde dehydrogenase (RALDH) 1 knockout (KO) mice fed a high-fat (HF) diet. FEN completely inhibited adipocyte differentiation by blocking CCAAT/enhancer-binding protein (C/EBP) α/peroxisome proliferator-activated receptor (PPAR) γ-mediated induction of downstream genes and upregulating RA-responsive genes like cellular retinol-binding protein-1. In mice fed an HF diet, RA-responsive genes were markedly increased in adipose, liver, and hypothalamus, with short-term and long-term FEN treatment. In adipose, FEN inhibited the downregulation of PPARγ and improved insulin sensitivity and the levels of adiponectin, resistin, and serum RBP (RBP4). FEN inhibited hyperleptinemia in vivo and leptin expression in adipocytes. Surprisingly, hypothalamic neuropeptide Y expression was completely suppressed, suggesting a central effect of FEN to normalize hyperglycemia. Moreover, FEN induced RA-responsive genes in RALDH1 KO mice, demonstrating that FEN can augment RA signaling when RA synthesis is impaired. We show that FEN-mediated beneficial effects are through alterations in retinoid homeostasis genes, and these are strong candidates as therapeutic targets for the treatment of obesity and insulin resistance.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Fármacos Antiobesidad/uso terapéutico , Fenretinida/uso terapéutico , Hipotálamo/efectos de los fármacos , Hígado/efectos de los fármacos , Obesidad/prevención & control , Retinoides/metabolismo , Células 3T3-L1 , Adipogénesis/efectos de los fármacos , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Fármacos Antiobesidad/farmacología , Dieta Alta en Grasa/efectos adversos , Fenretinida/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , ARN Mensajero/metabolismo , Distribución Aleatoria , Elementos de Respuesta/efectos de los fármacos , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/metabolismo , Proteínas Plasmáticas de Unión al Retinol/genética , Proteínas Plasmáticas de Unión al Retinol/metabolismo
5.
Leukemia ; 26(10): 2260-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22475870

RESUMEN

The in vitro effects of the synthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR, fenretinide) on primary B-cell chronic lymphocytic leukemia (CLL) cells from previously untreated CLL patients were investigated. 4HPR promoted the intrinsic apoptotic pathway by reactive oxygen species (ROS) generation and was accompanied by drop of Mcl-1 protein expression. The latter was not attributable to transcriptional downregulation but to protein degradation mediated by jun N-terminal kinase activation, and likely by NF-kB downregulation and Noxa upregulation. CLL cells stimulated in vitro with CD40L did not increase 4HPR chemoresistance if activation was accompanied by proliferation. Intra-patient analysis confirmed that the proliferating pool of CLL cells was more sensitive to the cytotoxic action of 4HPR than the activated but resting CLL subpopulation. The different 4HPR susceptibility of the two subpopulations was associated with higher Noxa expression in proliferating CLLs. Combination experiments revealed that 4HPR strongly potentiated ABT-737 cytotoxicity, especially in proliferating CLL cells that displayed amplified chemoresistance to ABT-737 alone. Synergic cytotoxicity was also demonstrated in combination with fludarabine, in both resting and stimulated CLL samples. This study entitles 4HPR to be assayed as a chemotherapeutic adjuvant for the treatment of CLL.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Fenretinida/farmacología , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Nitrofenoles/farmacología , Sulfonamidas/farmacología , Vidarabina/análogos & derivados , Proliferación Celular , Sinergismo Farmacológico , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Vidarabina/farmacología
6.
PLoS One ; 6(12): e28558, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22174837

RESUMEN

Glutathione (GSH) is implicated in drug resistance mechanisms of several cancers and is a key regulator of cell death pathways within cells. We studied Ewing's sarcoma family of tumours (ESFT) cell lines and three mechanistically distinct anticancer agents (fenretinide, doxorubicin, and vincristine) to investigate whether the GSH antioxidant system is involved in the reduced sensitivity to these chemotherapeutic agents in hypoxia. Cell viability and death were assessed by the trypan blue exclusion assay and annexin V-PI staining, respectively. Hypoxia significantly decreased the sensitivity of all ESFT cell lines to fenretinide-induced death, whereas the effect of doxorubicin or vincristine was marginal and cell-line-specific. The response of the GSH antioxidant system in ESFT cell lines to hypoxia was variable and also cell-line-specific, although the level of GSH appeared to be most dependent on de novo biosynthesis rather than recycling. RNAi-mediated knockdown of key GSH regulatory enzymes γ-glutamylcysteine synthetase or glutathione disulfide reductase partially reversed the hypoxia-induced resistance to fenretinide, and increasing GSH levels using N-acetylcysteine augmented the hypoxia-induced resistance in a cell line-specific manner. These observations are consistent with the conclusion that the role of the GSH antioxidant system in modulating the sensitivity of ESFT cells to fenretinide is heterogeneous depending on environment and cell type. This is likely to limit the value of targeting GSH as a therapeutic strategy to overcome hypoxia-induced drug resistance in ESFT. Whether targeting the GSH antioxidant system in conjunction with other therapeutics may benefit some patients with ESFT remains to be seen.


Asunto(s)
Antioxidantes/metabolismo , Fenretinida/uso terapéutico , Glutatión/metabolismo , Sarcoma de Ewing/tratamiento farmacológico , Sarcoma de Ewing/patología , Acetilcisteína/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Recuento de Células , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Suplementos Dietéticos , Ensayos de Selección de Medicamentos Antitumorales , Fenretinida/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Concentración 50 Inhibidora , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Modelos Biológicos , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sarcoma de Ewing/enzimología
7.
Cell Mol Life Sci ; 67(5): 807-16, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19941060

RESUMEN

Fenretinide induces apoptosis in neuroblastoma by induction of reactive oxygen species (ROS). In this study, we investigated the role of mitochondria in fenretinide-induced cytotoxicity and ROS production in six neuroblastoma cell lines. ROS induction by fenretinide was of mitochondrial origin, demonstrated by detection of superoxide with MitoSOX, the scavenging effect of the mitochondrial antioxidant MitoQ and reduced ROS production in cells without a functional mitochondrial respiratory chain (Rho zero cells). In digitonin-permeabilized cells, a fenretinide concentration-dependent decrease in ATP synthesis and substrate oxidation was observed, reflecting inhibition of the mitochondrial respiratory chain. However, inhibition of the mitochondrial respiratory chain was not required for ROS production. Co-incubation of fenretinide with inhibitors of different complexes of the respiratory chain suggested that fenretinide-induced ROS production occurred via complex II. The cytotoxicity of fenretinide was exerted through the generation of mitochondrial ROS and, at higher concentrations, also through inhibition of the mitochondrial respiratory chain.


Asunto(s)
Transporte de Electrón/efectos de los fármacos , Fenretinida/farmacología , Mitocondrias/efectos de los fármacos , Neuroblastoma/patología , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos/farmacología , Antioxidantes/farmacología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Humanos , Mitocondrias/metabolismo , Modelos Biológicos , Neuroblastoma/metabolismo , Compuestos Organofosforados/farmacología , Células Tumorales Cultivadas , Ubiquinona/análogos & derivados , Ubiquinona/farmacología
8.
Apoptosis ; 14(7): 849-63, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19421858

RESUMEN

Prostate tumorigenesis is coupled with an early metabolic switch in transformed prostate epithelial cells that effectively increases their mitochondrial bioenergetic capacity. The synthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) inhibits prostate cancer development in vivo, and triggers reactive oxygen species (ROS)-dependent prostate cancer cell apoptosis in vitro. The possibility that 4HPR-induced ROS production is associated with mitochondrial bioenergetics and required for apoptosis induction in transformed prostate epithelial cells in vitro would advocate a prospective mechanistic basis for 4HPR-mediated prostate cancer chemoprevention in vivo. We investigated this tenet by comparing and contrasting 4HPR's effects on premalignant PWR-1E and malignant DU-145 human prostate epithelial cells. 4HPR promoted a dose- and/or time-dependent apoptosis induction in PWR-1E and DU-145 cells, which was preceded by and dependent on an increase in mitochondrial ROS production. In this regard, the PWR-1E cells were more sensitive than the DU-145 cells, and they consumed roughly twice as much oxygen as the DU-145 cells suggesting oxidative phosphorylation was higher in the premalignant cells. Interestingly, increasing the [Ca(2+)] in the culture medium of the PWR-1E cells attenuated their proliferation as well as their mitochondrial bioenergetic capacity and 4HPR's cytotoxic effects. Correspondingly, the respiration-deficient derivatives (i.e., rho(0) cells lacking mitochondrial DNA) of DU-145 cells were markedly resistant to 4HPR-induced ROS production and apoptosis. Together, these observations implied that the reduction of mitochondrial bioenergetics protected PWR-1E and DU-145 cells against the cytotoxic effects of 4HPR, and support the concept that oxidative phosphorylation is an essential determinant in 4HPR's apoptogenic signaling in transformed human prostate epithelial cells.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Células Epiteliales , Fenretinida/farmacología , Mitocondrias , Próstata , Neoplasias de la Próstata , Animales , Ciclo Celular/fisiología , Línea Celular/efectos de los fármacos , Línea Celular/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Consumo de Oxígeno , Próstata/citología , Próstata/efectos de los fármacos , Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Especies Reactivas de Oxígeno/metabolismo
9.
BMC Cancer ; 9: 97, 2009 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-19331667

RESUMEN

BACKGROUND: Resistance of high-risk metastatic neuroblastoma (HR-NB) to high dose chemotherapy (HD-CT) raises a major therapeutic challenge in pediatric oncology. Patients are treated by maintenance CT. For some patients, an adjuvant retinoid therapy is proposed, such as the synthetic retinoid fenretinide (4-HPR), an apoptotic inducer. Recent studies demonstrated that NB metastasis process is enhanced by the loss of caspase-8 involved in the Integrin-Mediated Death (IMD) process. As the role of caspase-8 appears to be critical in preventing metastasis, we aimed at studying the effect of 4-HPR on caspase-8 expression in metastatic neuroblasts. METHODS: We used the human IGR-N-91 MYCN-amplified NB experimental model, able to disseminate in vivo from the primary nude mouse tumor xenograft (PTX) into myocardium (Myoc) and bone marrow (BM) of the animal. NB cell lines, i.e., IGR-N-91 and SH-EP, were treated with various doses of Fenretinide (4-HPR), then cytotoxicity was analyzed by MTS proliferation assay, apoptosis by the propidium staining method, gene or protein expressions by RT-PCR and immunoblotting and caspases activity by colorimetric protease assays. RESULTS: The IGR-N-91 parental cells do not express detectable caspase-8. However the PTX cells established from the primary tumor in the mouse, are caspase-8 positive. In contrast, metastatic BM and Myoc cells show a clear down-regulation of the caspase-8 expression. In parallel, the caspases -3, -9, -10, Bcl-2, or Bax expressions were unchanged. Our data show that in BM, compared to PTX cells, 4-HPR up-regulates caspase-8 expression that parallels a higher sensitivity to apoptotic cell death. Stable caspase-8-silenced SH-EP cells appear more resistant to 4-HPR-induced cell death compared to control SH-EP cells. Moreover, 4-HPR synergizes with drugs since apoptosis is restored in VP16- or TRAIL-resistant-BM cells. These results demonstrate that 4-HPR in up-regulating caspase-8 expression, restores and induces apoptotic cell death in metastatic neuroblasts through caspase-8 activation. CONCLUSION: This study provides basic clues for using fenretinide in clinical treatment of HR-NB patients. Moreover, since 4-HPR induces cell death in caspase-8 negative NB, it also challenges the concept of including 4-HPR in the induction of CT of these patients.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasa 8/metabolismo , Fenretinida/farmacología , Neuroblastoma/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Antineoplásicos/farmacología , Western Blotting , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Médula Ósea/patología , Caspasa 8/genética , Inhibidores de Caspasas , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Etopósido/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Neuroblastoma/enzimología , Neuroblastoma/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
10.
Cancer Res ; 68(13): 5363-9, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18593938

RESUMEN

Exploiting vulnerabilities in the intracellular signaling pathways of tumor cells is a key strategy for the development of new drugs. The activation of cellular stress responses mediated by the endoplasmic reticulum (ER) allows cancer cells to survive outside their normal environment. Many proteins that protect cells against ER stress are active as protein disulfide isomerases (PDI) and the aim of this study was to test the hypothesis that apoptosis in response to ER stress can be increased by inhibiting PDI activity. We show that the novel chemotherapeutic drugs fenretinide and velcade induce ER stress-mediated apoptosis in melanoma cells. Both stress response and apoptosis were enhanced by the PDI inhibitor bacitracin. Overexpression of the main cellular PDI, procollagen-proline, 2-oxoglutarate-4-dioxygenase beta subunit (P4HB), resulted in increased PDI activity and abrogated the apoptosis-enhancing effect of bacitracin. In contrast, overexpression of a mutant P4HB lacking PDI activity did not increase cellular PDI activity or block the effects of bacitracin. These results show that inhibition of PDI activity increases apoptosis in response to agents which induce ER stress and suggest that the development of potent, small-molecule PDI inhibitors has significant potential as a powerful tool for enhancing the efficacy of chemotherapy in melanoma.


Asunto(s)
Apoptosis/efectos de los fármacos , Bacitracina/farmacología , Retículo Endoplásmico/efectos de los fármacos , Melanoma/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bacitracina/administración & dosificación , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/farmacología , Bortezomib , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Fenretinida/administración & dosificación , Fenretinida/farmacología , Humanos , Melanoma/patología , Estrés Oxidativo/fisiología , Pirazinas/administración & dosificación , Pirazinas/farmacología , Resultado del Tratamiento , Células Tumorales Cultivadas
11.
Carcinogenesis ; 28(2): 378-89, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16950796

RESUMEN

O6-methylguanine-DNA methyltransferase (MGMT) is a DNA repair protein which protects the cellular genome and critical oncogenic genes from the mutagenic action of endogenous and exogenous alkylating agents. An expedited elimination of O6-alkylguanines by increasing MGMT activity levels is likely to be a successful chemoprevention strategy. Here, we report for the first time that cysteine/glutathione enhancing drugs and certain plant antioxidants possess the ability to increase human MGMT expression beyond its steady-state levels that may afford protection. The non-toxic cysteine prodrugs, 2-oxothiazolidine-4-carboxylic acid (OTC) and N-acetyl-L-cysteine (NAC), metabolized, respectively by 5-oxoprolinase and acylases, increased the MGMT protein and its repair activity levels in a dose- and time-dependent manner in several cancer cell lines and peripheral blood lymphocytes with a maximum of 3-fold increase by 72 h. The natural antioxidants, namely, curcumin, silymarin, sulforaphane and resveratrol were also effective in raising the MGMT levels to different extents. Among the synthetic agents, oltipraz and N-(4-hydroxyphenyl) retinamide (4-HPR) also increased MGMT expression, albeit to a lesser extent. Augmented mRNA levels accounted at least, in part, for the increased activity of MGMT in this setting. However, evidence from cysteine/methionine deprivation, acivicin treatment, and protein synthesis measurements in OTC-treated cells suggested that an increased cysteine flux also contributed significantly to enhanced MGMT expression. Many of these treatments increased the glutathione S-transferase-P1 (GSTP1) levels as well. These findings raise the possibility of MGMT-targeted chemoprevention strategies through dietary supplementation of OTC and herbal antioxidants. Further, the studies reveal the antioxidant responsiveness of the human MGMT gene.


Asunto(s)
Anticarcinógenos/farmacología , Productos Biológicos/farmacología , Cisteína/química , Linfocitos/metabolismo , Profármacos/farmacología , Proteína p14ARF Supresora de Tumor/genética , Acetilcisteína/farmacología , Antioxidantes/farmacología , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Curcumina/farmacología , Metilasas de Modificación del ADN , Cartilla de ADN , Enzimas Reparadoras del ADN , Relación Dosis-Respuesta a Droga , Fenretinida/farmacología , Glutatión/metabolismo , Gutatión-S-Transferasa pi/metabolismo , Humanos , Profármacos/química , Pirazinas/farmacología , ARN Mensajero/genética , Silimarina/farmacología , Tionas , Tiofenos , Proteína p14ARF Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor
12.
Clin Cancer Res ; 12(3 Pt 1): 969-79, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16467112

RESUMEN

Fenretinide [N-(4-hydroxyphenyl)retinamide or 4-HPR] is a synthetic retinoid analogue with antitumor and chemopreventive activities. N-(4-Methoxyphenyl)retinamide (4-MPR) is the most abundant metabolite of 4-HPR detected in human serum following 4-HPR therapy. We have shown in in vitro studies that 4-HPR and 4-MPR can act independent of the classic nuclear retinoid receptor pathway and that 4-HPR, but not 4-MPR, can also activate nuclear retinoid receptors. In this study, we have compared the chemopreventive effects of topically applied 4-HPR and 4-MPR with the primary biologically active retinoid, all-trans retinoic acid (ATRA), in vivo in the mouse skin two-stage chemical carcinogenesis model. All three retinoids suppressed tumor formation but the effect of 4-HPR and 4-MPR, and not of ATRA, was sustained after their discontinuation. The tumor-suppressive effects of 4-HPR and 4-MPR were quantitatively and qualitatively similar, suggesting that the two may be acting through the same retinoid receptor-independent mechanism(s). We further explored this effect in vitro by analyzing primary cultures of mouse keratinocytes treated with the same retinoids. All three could induce apoptosis with a 48-hour treatment and only ATRA and 4-HPR induced an accumulation of cells in the G1 phase of the cell cycle. This finding is consistent with our previous results showing that the effects of phenylretinamides on the cell cycle are retinoid receptor dependent whereas apoptosis induction is not. A microarray-based comparison of gene expression profiles for mouse skin treated with 12-O-tetradecanoylphorbol-13-acetate (TPA) alone and TPA + 4-HPR or TPA + 4-MPR reveals a high degree of coincidence between the genes regulated by the two phenylretinamides. We propose that 4-HPR may exert therapeutic and chemopreventive effects by acting primarily through a retinoid receptor-independent mechanism(s) and that 4-MPR may contribute to the therapeutic effect of 4-HPR by acting through the same retinoid receptor-independent mechanism(s).


Asunto(s)
Antineoplásicos/farmacología , Fenretinida/farmacología , Receptores X Retinoide/efectos de los fármacos , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/prevención & control , Tretinoina/análogos & derivados , Proteínas Supresoras de Tumor/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Quimioprevención , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Fase G1/efectos de los fármacos , Perfilación de la Expresión Génica , Técnicas In Vitro , Queratinocitos/efectos de los fármacos , Ratones , Ratones Endogámicos SENCAR , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Cutáneas/inducido químicamente , Acetato de Tetradecanoilforbol/farmacología , Tretinoina/farmacología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
13.
Oncogene ; 24(54): 7976-83, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16091745

RESUMEN

Fenretinide (4-HPR) is a synthetic retinoid whose apoptosis-inducing effects have been demonstrated in many tumor types. The precise mechanism of its apoptotic action is not fully understood. To further study the mechanism by which 4-HPR exerts its biological effects in neuroblastoma (NB) and to identify the genes that contribute to the induction of apoptosis, we determined the sensitivity of eight NB cell lines to 4-HPR. Additionally, cDNA microarray analysis was performed on a 4-HPR-sensitive cell line to investigate the temporal changes in gene expression, primarily focusing on the induction of proapoptotic genes. BBC3, a transcriptionally regulated proapoptotic member of the BCL2 family, was the most highly induced proapoptotic gene. Western analysis confirmed the induction of BBC3 protein by 4-HPR. Furthermore, the induction of BBC3 was associated with the sensitivity to this agent in the cell lines tested. Finally we demonstrated that BBC3 alone is sufficient to induce cell death in the 4-HPR-sensitive and resistant NB cell lines, and that siRNA against BBC3 significantly decreases apoptosis induced by 4-HPR. Our results indicate that BBC3 mediates cell death in NB cells in response to 4-HPR.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Fenretinida/farmacología , Proteínas de Neoplasias/fisiología , Neuroblastoma/tratamiento farmacológico , Proteínas Proto-Oncogénicas/metabolismo , Western Blotting , División Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Colorimetría , ADN Complementario , ADN de Neoplasias/análisis , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Cinética , Análisis por Micromatrices , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba/efectos de los fármacos
14.
Blood ; 103(9): 3516-20, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-14695237

RESUMEN

The anti-CD20 monoclonal antibody rituximab induces remission in 40% to 60% of patients with indolent B-cell lymphoma, but virtually all patients have relapses. We evaluated the efficacy of concurrent administration of another biologic agent, N-(4-hydroxyphenyl) retinamide (4HPR, fenretinide) with rituximab against a variety of human B-cell lymphoma cell lines (Ramos, DHL-4, and FL-18) in vivo. Concurrent 4HPR and rituximab administration prevented tumor progression of lymphoma-bearing mice in a minimal disease model (rituximab + 4HPR, 100% progression free; rituximab alone, 37.5% progression free, P =.01; 4HPR alone, 12.5% progression free, P <.01; controls, 0% progression free, P <.01). Combinations of 4HPR + rituximab exceeded the predicted 50% additive rate of disease control from each agent alone (P =.038). Administering 4HPR and rituximab to mice with established tumors induced complete responses (CRs) in 80% of animals compared with 20% to 40% CRs using either agent alone (P =.07), resulting in significantly improved survival. Tumors harvested from 4HPR + rituximab-treated mice displayed elevated caspase activation compared with untreated controls (P =.02). Adding a broad-spectrum caspase inhibitor in vivo fully abrogated the antitumor effects of 4HPR + rituximab (P =.05). These results establish the efficacy of 4HPR/rituximab combinations, confirm their caspase-mediated mechanism of action, and offer the potential for disease control with minimal toxicity for patients with B-cell malignancies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Fenretinida/farmacología , Linfoma de Células B/tratamiento farmacológico , Animales , Anticuerpos Monoclonales de Origen Murino , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Caspasas/metabolismo , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Femenino , Humanos , Linfoma de Células B/patología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasia Residual/tratamiento farmacológico , Inducción de Remisión/métodos , Rituximab , Tasa de Supervivencia , Trasplante Heterólogo
15.
Expert Opin Investig Drugs ; 12(11): 1829-42, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14585058

RESUMEN

Fenretinide (N-4-hydroxyphenylretinamide [4-HPR]) is a synthetic retinoid that has been examined in in vitro assays, preclinical animal models and clinical trials as a cancer chemopreventive agent. Its pharmacology, toxicity and mechanisms of action initially suggested an increased therapeutic index relative to native retinoids for the control of tumours of the breast, prostate, bladder, colon, cervix and head and neck. Although fenretinide at the doses and schedules used in several pivotal Phase II and III clinical trials has not been proven to be efficacious in reducing the incidence of cancer or in retarding the development of preneoplastic lesions, encouraging observations regarding unanticipated preventative activity, such as for ovarian cancer control, have arisen from these studies. Research in cancer therapy and the elucidation of molecular pathways activated by fenretinide have also yielded clues about how this agent might be better used in a prevention setting. Current trials are underway to re-examine both dose and schedule of fenretinide administration as well as the target tissues of interest. Investigations of potential synergism between fenretinide and other candidate chemopreventative molecules with complementary mechanisms of action may support future assessments of this prototype cancer prevention drug or its newer analogues.


Asunto(s)
Anticarcinógenos/uso terapéutico , Fenretinida/uso terapéutico , Neoplasias/prevención & control , Animales , Anticarcinógenos/efectos adversos , Anticarcinógenos/farmacología , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Fenretinida/efectos adversos , Fenretinida/farmacología , Humanos , Lesiones Precancerosas/prevención & control
16.
Int J Biol Markers ; 18(1): 70-4, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12699068

RESUMEN

Tumors growing within the host form dynamic aberrant tissue that consists of host components, including the stroma, an expanding vasculature and often chronic inflammation, in addition to the tumor cells themselves. These host components can contribute to, rather than limit, tumor expansion, whereas deprivation of vessel formation has the potential to confine tumors in small, clinically silent foci. Therapeutic inhibition of vessel formation could be best suited to preventive strategies aimed at the suppression of angiogenesis in primary tumors in subjects at risk, or of micrometastases after surgical removal of a primary tumor. Our analysis of potential cancer chemopreventive molecules including N-acetylcysteine, green tea flavonoids and 4-hydroxyphenyl-retinamide has identified antiangiogenic activities that could account--at least in part--for the tumor prevention effects observed with these compounds. These drugs appear to target common mechanisms of tumor angiogenesis that may permit identification of critical targets for antiangiogenic therapy and antiangiogenic chemoprevention.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticarcinógenos/farmacología , Catequina/análogos & derivados , Fenretinida/farmacología , Flavonoides/farmacología , Neoplasias/prevención & control , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Catequina/farmacología , Quimiotaxis , Endotelio Vascular/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/prevención & control , , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Int J Cancer ; 104(5): 559-67, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12594810

RESUMEN

Neuroblastoma (NB) is the most common extracranial solid tumor of childhood. In advanced disease stages, prognosis is poor and treatments have limited efficacy, thus novel strategies are warranted. The synthetic retinoid Fenretinide (HPR) induces apoptosis in NB and melanoma cell lines. We reported an in vitro potentiation of HPR effects on melanoma cells when the drug is incorporated into GD2-targeted immunoliposomes (anti-GD2-SIL-HPR). We investigated the antitumor activity of anti-GD2-SIL-HPR against NB cells, both in vitro and in vivo. Anti-GD2-SIL showed specific, competitive binding to and uptake by, various NB cell lines. In in vitro cytotoxicity studies, NB cells, incubated with 30 microM HPR entrapped in anti-GD2-immunoliposomes, showed a significant reduction in cellular growth compared to free HPR, HPR entrapped in Ab-free liposomes or anti-GD2 empty liposomes. In an in vivo NB metastatic model, we demonstrated that anti-GD2-SIL-HPR completely inhibited the development of macroscopic and microscopic metastases in comparison to controls. Similar, but significantly less potent, antitumor effect was observed also in mice treated with anti-GD2 immunoliposomes without HPR (anti-GD2-SIL-blank) or anti-GD2 MAb alone (p = 0.0297 and p = 0.0294, respectively, vs. anti-GD2-SIL-HPR). Moreover, our results clearly demonstrated that although anti-GD2 MAb had a strong antitumor effect in this in vivo NB model, 100% curability was obtained only after treatment with anti-GD2-SIL-HPR (p < 0.0001). Anti-GD2 liposomal HPR should receive clinical evaluation as adjuvant therapy of neuroblastoma.


Asunto(s)
Apoptosis/efectos de los fármacos , Fenretinida/farmacología , Fenretinida/uso terapéutico , Liposomas/administración & dosificación , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Animales , División Celular , Modelos Animales de Enfermedad , Fenretinida/administración & dosificación , Humanos , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Trasplante de Neoplasias , Análisis de Supervivencia , Factores de Tiempo , Células Tumorales Cultivadas
18.
Leuk Res ; 27(3): 259-66, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12537979

RESUMEN

BACKGROUND: Retinoids have been shown to regulate vital cellular processes including cell proliferation, differentiation and apoptosis. N-(4-Hydroxyphenyl)-all-trans-retinamide (fenretinide, 4-HPR) is a synthetic ATRA derivative with chemopreventive and cytotoxic activity against various cancer cell lines including myeloid leukemia. Although several modes of action have been postulated, its mechanism of action in hematologic malignancies remains unclear. Furthermore, only limited information exists as to its activity in lymphoid malignancies. METHODS AND RESULTS: To test whether 4-HPR has activity in acute lymphoblastic leukemia (ALL), we first analyzed its antiproliferative effect in five ALL (Z-33, Z-138, Z-119, Z-181, and Jurkat) cell lines. We found that 4-HPR inhibited the proliferation of all cell lines in a dose-dependent manner at concentrations ranging from 1 to 10 microM. We further demonstrated by cell cycle analysis that 5 microM of 4-HPR blocked Z-119 cells in S phase thus preventing their progression through the cycle. Next we tested whether 4-HPR activated the caspase pathway and induced apoptotic cell death. We found that 4-HPR induced apoptosis in Z-119 cells through the activation of caspase-3 and subsequent cleavage of its substrate poly(ADP-ribose) polymerase (PARP). We then asked whether 4-HPR could affect fresh ALL progenitor cells. Therefore, we obtained bone marrow and peripheral blood cells from five patients with newly diagnosed ALL and tested the effect of 4-HPR using the ALL blast colony culture assay. To supplement our results, we also performed the ALL blast assay on one ALL cell line (ALL-1). We found that 4-HPR significantly inhibited ALL colony-forming cell proliferation in a dose-dependent manner. CONCLUSIONS: Our data show that 4-HPR is a potent inhibitor of ALL cell proliferation and that it induces in vitro apoptotic cell death in ALL blasts. Further studies are warranted to establish the in vivo effect of 4-HPR particularly in patients with ALL.


Asunto(s)
Anticarcinógenos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Fenretinida/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Células Sanguíneas/patología , Células de la Médula Ósea/patología , Caspasa 3 , Caspasas/metabolismo , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Interfase/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Retinoides/farmacología , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
19.
Anticancer Res ; 21(3B): 1691-5, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11497248

RESUMEN

The expression of retinoid receptors is altered during the development of several types of cancer. In the present study, we determined the influence of high dietary concentrations of 4-hydroxyphenylretinamide (4-HPR) and 13-cis-retinoic acid (13-cis-RA) on RAR-beta mRNA expression in female mice. Expression of liver and lung RAR-beta RNA increased with increasing levels of dietary retinoid (both 4-HPR and 13-cis RA). Bladder RAR-beta mRNA levels, however, were significantly decreased in mice fed 13-cis RA or 4-HPR. These results suggest that feeding high levels of retinoids to mice results in tissue-specific elfects on expression of RAR-beta mRNA.


Asunto(s)
Anticarcinógenos/farmacología , Fenretinida/farmacología , Isotretinoína/farmacología , ARN Mensajero/metabolismo , Receptores de Ácido Retinoico/biosíntesis , Animales , Northern Blotting , ADN Complementario/metabolismo , Femenino , Hígado/metabolismo , Pulmón/metabolismo , Ratones , ARN/metabolismo , Distribución Tisular , Vejiga Urinaria/metabolismo
20.
Curr Oncol Rep ; 2(6): 511-8, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11122886

RESUMEN

Retinoids are derivatives of vitamin A that include all- trans-retinoic acid (ATRA), 13-cis-retinoic acid, (13-cis-RA), and fenretinide (4-HPR). High levels of either ATRA or 13-cis-RA can cause arrest of cell growth and morphologic differentiation of human neuroblastoma cell lines. Phase I trials have shown that higher and more sustained drug levels were obtained with 13-cis-RA relative to ATRA. A phase III randomized trial showed that high-dose pulse therapy with 13-cis-RA given after completion of intensive chemoradiotherapy (with or without autologous bone marrow transplantation) significantly improves event-free survival in high-risk neuroblastoma. Because 4-HPR achieves multi-log cell kills in neuroblastoma cell lines that are resistant to ATRA and 13-cis-RA, a pediatric phase I trial is in progress to determine the maximum tolerated dose of 4-HPR, with a view toward giving 4-HPR after completion of myeloablative therapy and 13-cis-RA.


Asunto(s)
Antineoplásicos/farmacología , Fenretinida/farmacología , Isotretinoína/farmacología , Neuroblastoma/tratamiento farmacológico , Tretinoina/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Niño , Esquema de Medicación , Fenretinida/administración & dosificación , Fenretinida/efectos adversos , Humanos , Isotretinoína/administración & dosificación , Isotretinoína/efectos adversos , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptores de Ácido Retinoico/efectos de los fármacos , Receptores de Ácido Retinoico/fisiología , Tretinoina/administración & dosificación , Tretinoina/efectos adversos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA