Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Environ Res ; 189: 109845, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32678729

RESUMEN

Detroit, Michigan, currently has the highest preterm birth (PTB) rate of large cities in the United States. Disproportionate exposure to ambient air pollutants, including particulate matter ≤2.5 µm (PM2.5), PM ≤ 10 µm (PM10), nitrogen dioxide (NO2) and benzene, toluene, ethylbenzene, and xylenes (BTEX) may contribute to PTB. Our objective was to examine the association of airshed pollutants with PTB in Detroit, MI. The Geospatial Determinants of Health Outcomes Consortium (GeoDHOC) study collected air pollution measurements at 68 sites in Detroit in September 2008 and June 2009. GeoDHOC data were coupled with 2008-2010 Michigan Air Sampling Network measurements in Detroit to develop monthly ambient air pollution estimates at a spatial density of 300 m2. Using delivery records from two urban hospitals, we established a retrospective birth cohort of births by Detroit women occurring from June 2008 to May 2010. Estimates of air pollutant exposure throughout pregnancy were assigned to maternal address at delivery. Our analytic sample size included 7961 births; 891 (11.2%) were PTB. After covariate adjustment, PM10 (P = 0.003) and BTEX (P < 0.001), but not PM2.5 (P = 0.376) or NO2 (P = 0.582), were statistically significantly associated with PTB. In adjusted models, for every 5-unit increase in PM10 there was a 1.21 times higher odds of PTB (95% CI 1.07, 1.38) and for every 5-unit increase in BTEX there was a 1.54 times higher odds of PTB (95% CI 1.25, 1.89). Consistent with previous studies, higher PM10 was associated with PTB. We also found novel evidence that higher airshed BTEX is associated with PTB. Future studies confirming these associations and examining direct measures of exposure are needed.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire , Contaminantes Ambientales , Nacimiento Prematuro , Contaminantes Atmosféricos/análisis , Contaminantes Atmosféricos/toxicidad , Contaminación del Aire/efectos adversos , Ciudades , Estudios de Cohortes , Exposición a Riesgos Ambientales/análisis , Femenino , Humanos , Recién Nacido , Exposición Materna , Michigan/epidemiología , Material Particulado/análisis , Material Particulado/toxicidad , Embarazo , Nacimiento Prematuro/inducido químicamente , Nacimiento Prematuro/epidemiología , Estudios Retrospectivos
2.
Drug Metab Dispos ; 44(5): 665-71, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26953171

RESUMEN

CYP1A1 and CYP1A2 are transcriptionally activated in the human normal breast epithelial cell line MCF10A following exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Shifting MCF10A cultures to medium deficient in serum and epidermal growth factor (EGF) caused rapid reductions in the activated (i.e., phosphorylated) forms of extracellular regulated kinases (ERKs) and the epidermal growth factor receptor (EGFR). Shifting to serum/EGF-deficient medium also enhanced TCDD-mediated induction of cytochrome P450 (CYP)1A1 Treatment of cells cultured in complete medium with the EGFR inhibitors gefitinib (Iressa), AG1478, and CI-1033 resulted in concentration-dependent reductions of active EGFR and ERKs, and increased CYP1A1 mRNA content ∼3- to 18-fold above basal level. EGFR inhibitors synergized with TCDD and resulted in transient CYP1A1 and CYP1A2 mRNA accumulations ∼8-fold greater (maximum at 5 hours) than that achieved with only TCDD. AG1478, gefitinib, and TCDD individually induced small increases (∼1.2- to 2.5-fold) in CYP1A1 protein content but did not cause additive or synergistic accumulations of CYP1A1 protein when used in combination. The mitogen-activated protein kinase kinase inhibitor PD184352 inhibited ERK and EGFR activation in a concentration-dependent fashion without causing CYP1A1 mRNA accumulation. However, cotreatment with PD184352 potentiated TCDD-mediated CYP1A1 induction. TCDD-mediated induction of CYP1A1 in MCF7-TET on-EGFR cells, a MCF7 variant in which EGFR expression can be controlled, was not affected by the activity status of EGFR or ERKs. Hence, EGFR signaling mutes both basal and ligand-induced expression of two aryl hydrocarbon receptor-responsive P450s in MCF10A cultures. However, these effects are cell context-dependent. Furthermore, CYP1A1 mRNA and protein abundance are not closely coupled in MCF10A cultures.


Asunto(s)
Mama/efectos de los fármacos , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Células Epiteliales/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Dibenzodioxinas Policloradas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Benzamidas/farmacología , Mama/metabolismo , Línea Celular , Sinergismo Farmacológico , Células Epiteliales/metabolismo , Gefitinib , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Morfolinas/farmacología , Quinazolinas/farmacología , ARN Mensajero/metabolismo , Tirfostinos/farmacología
3.
Artículo en Inglés | MEDLINE | ID: mdl-38541258

RESUMEN

African American women in the United States have a high risk of adverse pregnancy outcomes. DNA methylation is a potential mechanism by which exposure to BTEX (benzene, toluene, ethylbenzene, and xylenes) may cause adverse pregnancy outcomes. Data are from the Maternal Stress Study, which recruited African American women in the second trimester of pregnancy from February 2009 to June 2010. DNA methylation was measured in archived DNA from venous blood collected in the second trimester. Trimester-specific exposure to airshed BTEX was estimated using maternal self-reported addresses and geospatial models of ambient air pollution developed as part of the Geospatial Determinants of Health Outcomes Consortium. Among the 64 women with exposure and outcome data available, 46 differentially methylated regions (DMRs) were associated with BTEX exposure (FDR adjusted p-value < 0.05) using a DMR-based epigenome-wide association study approach. Overall, 89% of DMRs consistently exhibited hypomethylation with increasing BTEX exposure. Biological pathway analysis identified 11 enriched pathways, with the top 3 involving gamma-aminobutyric acid receptor signaling, oxytocin in brain signaling, and the gustation pathway. These findings highlight the potential impact of BTEX on DNA methylation in pregnant women.


Asunto(s)
Contaminantes Atmosféricos , Benceno , Negro o Afroamericano , Metilación de ADN , Femenino , Humanos , Embarazo , Contaminantes Atmosféricos/toxicidad , Contaminantes Atmosféricos/análisis , Benceno/análisis , Benceno/toxicidad , Derivados del Benceno/análisis , Derivados del Benceno/toxicidad , Negro o Afroamericano/genética , Monitoreo del Ambiente , Tolueno/toxicidad , Tolueno/análisis , Xilenos/toxicidad , Xilenos/análisis
4.
Cancers (Basel) ; 16(1)2023 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-38201517

RESUMEN

Neurofibromatosis type 1 (NF1) is a disorder in which RAS is constitutively activated due to the loss of the Ras-GTPase-activating activity of neurofibromin. RAS must be prenylated (i.e., farnesylated or geranylgeranylated) to traffic and function properly. Previous studies showed that the anti-growth properties of farnesyl monophosphate prodrug farnesyltransferase inhibitors (FTIs) on human NF1 malignant peripheral nerve sheath tumor (MPNST) cells are potentiated by co-treatment with lovastatin. Unfortunately, such prodrug FTIs have poor aqueous solubility. In this study, we synthesized a series of prodrug FTI polyamidoamine generation 4 (PAMAM G4) dendrimers that compete with farnesyl pyrophosphate for farnesyltransferase (Ftase) and assessed their effects on human NF1 MPNST S462TY cells. The prodrug 3-tert-butylfarnesyl monophosphate FTI-dendrimer (i.e., IG 2) exhibited improved aqueous solubility. Concentrations of IG 2 and lovastatin (as low as 0.1 µM) having little to no effect when used singularly synergistically suppressed cell proliferation, colony formation, and induced N-RAS, RAP1A, and RAB5A deprenylation when used in combination. Combinational treatment had no additive or synergistic effects on the proliferation/viability of immortalized normal rat Schwann cells, primary rat hepatocytes, or normal human mammary epithelial MCF10A cells. Combinational, but not singular, in vivo treatment markedly suppressed the growth of S462TY xenografts established in the sciatic nerves of immune-deficient mice. Hence, prodrug farnesyl monophosphate FTIs can be rendered water-soluble by conjugation to PAMAM G4 dendrimers and exhibit potent anti-tumor activity when combined with clinically achievable statin concentrations.

5.
Drug Metab Dispos ; 40(5): 1032-40, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22344700

RESUMEN

The aryl hydrocarbon receptor (AhR) is targeted by ubiquitination for degradation by the proteasome shortly after its activation by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). In silico screening identified p-anilinoaniline (pAA) as a putative inhibitor of an E2 ligase that partners with an E3 ligase implicated in AhR ubiquitination. We investigated whether pAA could modify AhR-dependent activation of its target gene CYP1A1. pAA (1-200 µM) alone did not affect AhR content, or stimulate CYP1A1 mRNA accumulation in human mammary epithelial MCF10A cultures. However, pretreatment with ≥100 µM pAA suppressed TCDD-induced CYP1A1 activation and AhR degradation via its functioning as an AhR antagonist. At a lower concentration (25 µM), pAA cotreatment increased TCDD-induced CYP1A1 mRNA accumulation, without inhibiting AhR turnover or altering CYP1A1 mRNA half-life. Whereas TCDD alone did not affect MCF10A proliferation, 25 µM pAA was cytostatic and induced a G(1) arrest that lasted ∼7 h and induced an S phase arrest that peaked 5 to 8 h later. TCDD neither affected MCF10A cell cycle progression nor did it alter pAA effects on the cell cycle. The magnitude of CYP1A1 activation depended upon the time elapsed between pAA pretreatment and TCDD addition. Maximal AhR occupancy of the CYP1A1 promoter and accumulation of CYP1A1 heterogeneous nuclear RNA and mRNA occurred when pAA-pretreated cultures were exposed to TCDD in late G(1) and early/mid S phase. TCDD-mediated induction of CYP2S1 was also cell cycle-dependent in MCF10A cultures. Similar studies with HepG2 cultures indicated that the cell cycle dependence of CYP1A1 induction is cell context-dependent.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Citocromo P-450 CYP1A1/metabolismo , Fenilendiaminas/farmacología , Dibenzodioxinas Policloradas/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Transcripción Genética , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1A1/genética , Ensayo de Cambio de Movilidad Electroforética , Inducción Enzimática , Humanos , Regiones Promotoras Genéticas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores
6.
J Pharmacol Exp Ther ; 337(1): 65-74, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21228063

RESUMEN

Exposure of the human malignant peripheral nerve sheath tumor cell lines STS-26T, ST88-14, and NF90-8 to nanomolar concentrations of both lovastatin and farnesyl transferase inhibitor (FTI)-1 but not to either drug alone induced cell death. ST88-14 and NF90-8 cells underwent apoptosis, yet dying STS-26T cells did not. FTI-1 cotreatment induced a strong and sustained autophagic response as indicated by analyses of microtubule-associated protein-1 light chain 3 (LC3)-II accumulation in STS-26T cultures. Extensive colocalization of LC3-positive punctate spots was observed with both lysosome-associated membrane protein (LAMP)-1 and LAMP-2 (markers of late endosomes/lysosomes) in solvent or FTI-1 or lovastatin-treated STS-26T cultures but very little colocalization in lovastatin/FTI-1-cotreated cultures. The absence of colocalization in the cotreatment protocol correlated with loss of LAMP-2 expression. Autophagic flux studies indicated that lovastatin/FTI-1 cotreatment inhibited the completion of the autophagic program. In contrast, rapamycin induced an autophagic response that was associated with cytostasis but maintenance of viability. These studies indicate that cotreatment of STS-26T cells with lovastatin and FTI-1 induces an abortive autophagic program and nonapoptotic cell death.


Asunto(s)
Apoptosis , Autofagia/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Farnesiltransferasa/antagonistas & inhibidores , Lovastatina/administración & dosificación , Animales , Autofagia/fisiología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Combinación de Medicamentos , Farnesiltransferasa/metabolismo , Humanos , Ratones , Ratas
7.
J Reprod Immunol ; 145: 103305, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33725526

RESUMEN

Air pollution is associated with preterm birth (PTB), potentially via inflammation. We recently showed the mixture benzene, toluene, ethylbenzene, and xylene (BTEX) is associated with PTB. We examined if ambient BTEX exposure is associated with mid-pregnancy inflammation in a sample of 140 African-American women residing in Detroit, Michigan. The Geospatial Determinants of Health Outcomes Consortium study collected outdoor air pollution measurements in Detroit; these data were coupled with Michigan Air Sampling Network measurements to develop monthly BTEX concentration estimates at a spatial density of 300 m2. First trimester and mid-pregnancy BTEX exposure estimates were assigned to maternal address. Mid-pregnancy (mean 21.3 ± 3.7 weeks gestation) inflammatory biomarkers (high-sensitivity C-reactive protein, interleukin [IL]-6, IL-10, IL-1ß, and tumor necrosis factor-α) were measured with enzyme immunoassays. After covariate adjustment, for every 1-unit increase in first trimester BTEX, there was an expected mean increase in log-transformed IL-1ß of 0.05 ± 0.02 units (P = 0.014) and an expected mean increase in log-transformed tumor necrosis factor-α of 0.07 ± 0.02 units (P = 0.006). Similarly, for every 1-unit increase in mid-pregnancy BTEX, there was a mean increase in log IL-1ß of 0.06 ± 0.03 units (P = 0.027). There was no association of either first trimester or mid-pregnancy BTEX with high-sensitivity C-reactive protein, IL-10, or IL-6 (all P > 0.05). Ambient BTEX exposure is associated with inflammation in mid-pregnancy in African-American women. Future studies examining if inflammation mediates associations between BTEX exposure and PTB are needed.


Asunto(s)
Contaminantes Atmosféricos/efectos adversos , Negro o Afroamericano/estadística & datos numéricos , Interleucina-1beta/sangre , Nacimiento Prematuro/inmunología , Factor de Necrosis Tumoral alfa/sangre , Adolescente , Adulto , Benceno/efectos adversos , Derivados del Benceno/efectos adversos , Biomarcadores/sangre , Exposición a Riesgos Ambientales/efectos adversos , Femenino , Humanos , Interleucina-1beta/inmunología , Exposición Materna/estadística & datos numéricos , Embarazo , Nacimiento Prematuro/sangre , Tolueno/efectos adversos , Factor de Necrosis Tumoral alfa/inmunología , Xilenos/efectos adversos , Adulto Joven
8.
J Pharmacol Exp Ther ; 333(1): 23-33, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20086055

RESUMEN

Prenylation inhibitors have gained increasing attention as potential therapeutics for cancer. Initial work focused on inhibitors of farnesylation, but more recently geranylgeranyl transferase inhibitors (GGTIs) have begun to be evaluated for their potential antitumor activity in vitro and in vivo. In this study, we have developed a nonpeptidomimetic GGTI, termed GGTI-2Z [(5-nitrofuran-2-yl)methyl-(2Z,6E,10E)-3,7,11,15-tetramethylhexadeca-2,6,10,14-tetraenyl 4-chlorobutyl(methyl)phosphoramidate], which in combination with lovastatin inhibits geranylgeranyl transferase I (GGTase I) and GGTase II/RabGGTase, without affecting farnesylation. The combination treatment results in a G(0)/G(1) arrest and synergistic inhibition of proliferation of cultured STS-26T malignant peripheral nerve sheath tumor cells. We also show that the antiproliferative activity of drugs in combination occurs in the context of autophagy. The combination treatment also induces autophagy in the MCF10.DCIS model of human breast ductal carcinoma in situ and in 1c1c7 murine hepatoma cells, where it also reduces proliferation. At the same time, there is no detectable toxicity in normal immortalized Schwann cells. These studies establish GGTI-2Z as a novel geranylgeranyl pyrophosphate derivative that may work through a new mechanism involving the induction of autophagy and, in combination with lovastatin, may serve as a valuable paradigm for developing more effective strategies in this class of antitumor therapeutics.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Antineoplásicos/farmacología , Autofagia , Diterpenos/farmacología , Lovastatina/farmacología , Compuestos Organofosforados/farmacología , Transferasas/antagonistas & inhibidores , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Fase G1/efectos de los fármacos , GTP Fosfohidrolasas/metabolismo , Humanos , Ratones , Prenilación de Proteína , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Células de Schwann/citología , Células de Schwann/efectos de los fármacos
9.
Mol Cell Biochem ; 344(1-2): 267-76, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20680410

RESUMEN

Plexiform neurofibromas commonly found in patients with Neurofibromatosis type I (NF1) have a 5% risk of being transformed into malignant peripheral nerve sheath tumors (MPNST). Germline mutations in the NF1 gene coding for neurofibromin, which is a Ras GTPase activating protein (RasGAP) and a negative regulator of Ras, result in an upregulation of the Ras pathway. We established a direct connection between neurofibromin deficiency and downstream effectors of Ras in cell lines from MPNST patients by demonstrating that knockdown of NF1 expression using siRNA in a NF1 wild type MPNST cell line, STS-26T, activates the Ras/ERK1,2 pathway and increases AP-1 binding and activity. We believe this is the first time the transactivation of AP-1 has been linked directly to neurofibromin deficiency in a disease relevant MPNST cell line. Previously, we have shown that N-Ras is constitutively activated in cell lines derived from independent MPNSTs from NF1 patients. We therefore sought to analyze the role of the N-Ras pathway in deregulating AP-1 transcriptional activity. We show that STS-26T clones conditionally expressing oncogenic N-Ras show increased phosphorylated ERK1,2 and phosphorylated JNK expression concomitant with increased AP-1 activity. MAP kinase pathways (ERK1,2 and JNK) were further examined in ST88-14, a neurofibromin-deficient MPNST cell line. The basal activity of ERK1,2 but not JNK was found to increase AP-1 activity. These experiments further confirmed the link between the loss of neurofibromin and increased activity of Ras/MAP kinase pathways and the activation of downstream transcriptional mechanisms in MPNSTs from NF1 patients.


Asunto(s)
Genes ras , Neoplasias de la Vaina del Nervio/fisiopatología , Neurofibromina 1/fisiología , Secuencia de Bases , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Neoplasias de la Vaina del Nervio/genética , Oligodesoxirribonucleótidos , Fosforilación
10.
Autophagy ; 16(11): 2098-2101, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32584644

RESUMEN

Macroautophagy/autophagy can play a cytoprotective role after photodynamic damage to malignant cells, depending on the site of subcellular damage initiated by reactive oxygen species. There is evidence for such protection when mitochondria are among the targets. Targeting lysosomes has been reported to be more effective for photokilling, perhaps because autophagy offers no cytoprotection. Photodynamic damage to both lysosomes and mitochondria can, however, markedly enhance the overall level of photokilling. Two mechanisms have been proposed to account for this result. Lysosomal photodamage leads to the release of calcium ions, resulting in the activation of the protease CAPN (calpain). CAPN then cleaves ATG5 to a fragment (tATG5) capable of interacting with mitochondria to enhance pro-apoptotic signals. It has also been proposed that targeting lysosomes for photodynamic damage can impair mitophagy, a process that could mitigate the pro-apoptotic effects of mitochondrial targeting. The level of lysosomal photodamage required for suppression of mitophagy is unclear. The "tATG5 route" involves the catalytic action of CAPN, activated by a degree of lysosomal photodamage barely detectible by a viability assay. ER photodamage can also initiate paraptosis, a death pathway functional even in cell types with impaired apoptosis and apparently unaffected by autophagy. Abbreviations: ALLN: N-acetyl-Leu-Leu-norleucinal (cell-permeable inhibitor of calpain); ATG: autophagy related; BPD: benzoporphyrin derivative (Visudyne); ER: endoplasmic reticulum; EtNBS: 5-ethylamino-9-diethyl-aminobenzo[a]phenothiazinium chloride; MTT: a tetrazolium dye; NPe6: mono N-aspartyl chlorin e6; PDT: photodynamic therapy; ROS: reactive oxygen species.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Retículo Endoplásmico/metabolismo , Mitofagia/fisiología , Fotoquimioterapia , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Lisosomas/metabolismo , Mitocondrias/metabolismo , Mitofagia/efectos de los fármacos , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo
11.
J Pharmacol Exp Ther ; 328(3): 796-806, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19066341

RESUMEN

The therapeutic usefulness of the quinoxaline derivatives XK469 (2-{4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy}propionic acid) and SH80 (2-{4-[(7-bromo-2-quinolinyl)oxy]phenoxy}propionic acid) has been attributed to their abilities to induce G(2)/M arrest and apoptotic or autophagic cell death. Concentrations of XK469 or SH80 > or = 5 microM were cytostatic to cultures of the normal murine melanocyte cell line Melan-a. Higher concentrations caused dose-dependent cytotoxicity. Concentrations > or =10 microM provoked dramatic morphological changes typified by marked increases in cell size and granularity. XK469/SH80-treated cultures accumulated tetraploid (4N) DNA-containing cells within 24 h of treatment, an 8N population within 3 days, and a 16N population within 5 days. Increases in ploidy correlated with the appearance of multinucleated cells. Under no circumstances did cells exhibit evidence of furrow formation. Both drugs suppressed cytokinesis in additional mammalian cell lines. Cytotoxic concentrations of XK469 elevated DEVDase activities (a measure of procaspase-3/7 activation) and enhanced cellular staining by a fluorescent analog of the pan caspase inhibitor valine-alanine-aspartic acid-fluoromethyl ketone within 48 to 96 h of treatment. Within 48 h of treatment, cytostatic and cytotoxic concentrations of XK469 elevated p21 contents, reduced Bcl-2 and Bcl-XL contents, and induced autophagy, as monitored by the accumulation of phosphatidylethanolamine-modified cleavage product of microtubule-associated protein light chain 3 (LC3-II). Cultures treated with > or =10 microM XK469 or SH80 for 5 days could not be induced to divide upon removal of drugs. Such cultures maintained high LC3-II contents, exhibited reduced cyclin E and D1 contents, and extensively expressed senescence-associated beta-galactosidase within 14 to 17 days of cessation of drug treatment. Hence, XK469 and SH80 inhibit cytokinesis, promote polyploidy, and induce senescence in Melan-a cells.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Citocinesis/efectos de los fármacos , Poliploidía , Propionatos/farmacología , Quinoxalinas/farmacología , Animales , Autofagia/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular/efectos de los fármacos , Células Cultivadas , Citometría de Flujo , Ratones , Péptido Hidrolasas/metabolismo
12.
Photochem Photobiol ; 85(5): 1177-81, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19508643

RESUMEN

Singlet oxygen (1O2) is the primary oxidant generated in photodynamic therapy (PDT) protocols involving sensitizers resulting in type II reactions. 1O2 can give rise to additional reactive oxygen species (ROS) such as the hydroxyl radical (*OH). The current study was designed to assess 3'-p-(aminophenyl) fluorescein (APF) and 3'-p-(hydroxyphenyl) fluorescein (HPF) as probes for the detection of 1O2 and *OH under conditions relevant to PDT. Cell-free studies indicated that both APF and HPF were converted to fluorescent products following exposure to 1O2 generated by irradiation of a water-soluble photosensitizing agent (TPPS) and that APF was 35-fold more sensitive than HPF. Using the 1O2 probe singlet oxygen sensor green (SOSG) we confirmed that 1 mm NaN3 quenched 1O2-induced APF/HPF fluorescence, while 1% DMSO had no effect. APF and HPF also yielded a fluorescent product upon interacting with *OH generated from H2O2 via the Fenton reaction in a cell-free system. DMSO quenched the fluorogenic interaction between APF/HPF and *OH at doses as low as 0.02%. Although NaN3 was expected to quench *OH-induced APF/HPF fluorescence, co-incubating NaN3 with APF or HPF in the presence of *OH markedly enhanced fluorescence. Cultured L1210 cells that had been photosensitized with benzoporphyhrin derivative exhibited APF fluorescence immediately following irradiation. Approximately 50% of the cellular fluorescence could be suppressed by inclusion of either DMSO or the iron-chelator desferroxamine. Combining the latter two agents did not enhance suppression. We conclude that APF can be used to monitor the formation of both 1O2 and *OH in cells subjected to PDT if studies are performed in the presence and absence of DMSO, respectively. That portion of the fluorescence quenched by DMSO will represent the contribution of *OH. This procedure could represent a useful means for evaluating formation of both ROS in the context of PDT.


Asunto(s)
Colorantes Fluorescentes/química , Radical Hidroxilo/metabolismo , Fotoquimioterapia , Oxígeno Singlete/metabolismo , Animales , Línea Celular Tumoral , Ratones , Espectrometría de Fluorescencia
13.
J Pharmacol Exp Ther ; 326(1): 1-11, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18367665

RESUMEN

Neurofibromatosis type 1 (NF1) is a genetic disorder that is driven by the loss of neurofibromin (Nf) protein function. Nf contains a Ras-GTPase-activating protein domain, which directly regulates Ras signaling. Numerous clinical manifestations are associated with the loss of Nf and increased Ras activity. Ras proteins must be prenylated to traffic and functionally localize with target membranes. Hence, Ras is a potential therapeutic target for treating NF1. We have tested the efficacy of two novel farnesyl transferase inhibitors (FTIs), 1 and 2, alone or in combination with lovastatin, on two NF1 malignant peripheral nerve sheath tumor (MPNST) cell lines, NF90-8 and ST88-14. Single treatments of 1, 2, or lovastatin had no effect on Ras prenylation or MPNST cell proliferation. However, low micromolar combinations of 1 or 2 with lovastatin (FTI/lovastatin) reduced Ras prenylation in both MPNST cell lines. Furthermore, this FTI/lovastatin combination treatment reduced cell proliferation and induced an apoptotic response as shown by morphological analysis, procaspase-3/-7 activation, loss of mitochondrial membrane potential, and accumulation of cells with sub-G(1) DNA content. Little to no detectable toxicity was observed in normal rat Schwann cells following FTI/lovastatin combination treatment. These data support the hypothesis that combination FTI plus lovastatin therapy may be a potential treatment for NF1 MPNSTs.


Asunto(s)
Apoptosis/efectos de los fármacos , Farnesiltransferasa/antagonistas & inhibidores , Lovastatina/administración & dosificación , Neoplasias de la Vaina del Nervio/tratamiento farmacológico , Neoplasias de la Vaina del Nervio/patología , Neurofibromatosis 1/tratamiento farmacológico , Neurofibromatosis 1/patología , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Línea Celular , Línea Celular Transformada , Línea Celular Tumoral , Quimioterapia Combinada , Inhibidores Enzimáticos/administración & dosificación , Farnesiltransferasa/metabolismo , Lovastatina/química , Neurofibromatosis 1/enzimología , Ratas , Ratas Sprague-Dawley
14.
Toxicol Appl Pharmacol ; 232(2): 169-79, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18655802

RESUMEN

p-Anilinoaniline (pAA) is an aromatic amine that is widely used in hair dying applications. It is also a metabolite of metanil yellow, an azo dye that is commonly used as a food coloring agent. Concentrations of pAA between 10 and 25 microM were cytostatic to cultures of the normal human mammary epithelia cell line MCF10A. Concentrations >or=50 microM were cytotoxic. Cytostatic concentrations induced transient G(1) and S cell cycle phase arrests; whereas cytotoxic concentrations induced protracted arrests. Cytotoxic concentrations of pAA caused DNA damage, as monitored by the alkaline single-cell gel electrophoresis (Comet) assay, and morphological changes consistent with cells undergoing apoptosis and/or autophagy. Enzymatic and western blot analyses, and binding analyses of fluorescent labeled VAD-FMK, suggested that caspase family members were activated by pAA. Western blot analyses documented the conversion of LC3-I to LC3-II, a post-translational modification involved in the development of the autophagosome. Suppression of autophagosome formation, via knockdown of ATG7 with shRNA, prevented pAA-induced vacuolization, enhanced the activation of pro-caspase-3, and increased susceptibility of ATG7-deficient cells to the cytostatic and cytotoxic activities of markedly lower concentrations of pAA. Cells stably transfected with a nonsense shRNA behaved like parental MCF10A cells. Collectively, these data suggest that MCF10A cultures undergo autophagy as a pro-survival response to concentrations of pAA sufficient to induce DNA damage.


Asunto(s)
Alanina/análogos & derivados , Autofagia/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Alanina/toxicidad , Aminoácidos Aromáticos/toxicidad , Apoptosis/efectos de los fármacos , Apoptosis/genética , Autofagia/genética , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Daño del ADN/fisiología , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Células Epiteliales/fisiología , Humanos
15.
Photochem Photobiol ; 84(5): 1272-6, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18513234

RESUMEN

The proapoptotic effects of the Bcl-2 antagonist HA14-1 are believed to derive from its affinity for the hydrophobic groove on Bcl-2 and Bcl-x(L), thereby displacing proapoptotic factors, e.g. Bax and Bak. We have reported that HA14-1 promotes the efficacy of low-dose photodynamic therapy (PDT). A recent report proposed that the proapoptotic activity of HA14-1 reflects its ability to generate reactive oxygen species (ROS) when incubated in an aqueous environment. This later study, like several other HA14-1 investigations, relied on the use of fluorescent probes for ROS detection. We found that HA14-1 reacts with the albumin in serum to yield a fluorescent product. After correcting for this effect, the putative formation of ROS by HA14-1 could not be demonstrated with the fluorescent probes H(2)DCFDA, dihydroethidium or dihydrorhodamine. Indeed, the fluorescence excitation/emission spectra of HA14-1 encompassed the excitation/emission wavelengths used to detect these ROS probes. Cells cultured in a medium supplemented with ovalbumin, instead of serum, underwent apoptosis following HA14-1 addition, but did not exhibit fluorescence. Hence, HA14-1 fluorescence was unrelated to its proapoptotic activity. We conclude that the enhancement of PDT by HA14-1 reflects a pharmacologic effect, rather than its direct contribution of ROS.


Asunto(s)
Benzopiranos/química , Fluorescencia , Colorantes Fluorescentes/química , Nitrilos/química , Fotoquimioterapia , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Especies Reactivas de Oxígeno/química , Albúmina Sérica/química , Animales , Apoptosis/efectos de los fármacos , Benzopiranos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Ratones , Nitrilos/farmacología , Oxidación-Reducción , Factores de Tiempo , Células Tumorales Cultivadas
16.
Mol Cancer Ther ; 6(1): 370-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17237296

RESUMEN

The phenoxypropionic acid derivative 2-{4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy}propionic acid (XK469) and an analogue termed 2-{4-[(7-bromo-2-quinalinyl)oxy]phenoxy}propionic acid (SH80) can eradicate malignant cell types resistant to many common antitumor agents. Colony formation assays indicated that a 24 h exposure of L1210 cells to XK469 or SH80 inhibited clonogenic growth with CI(90) values of 10 and 13 micromol/L, respectively. This effect was associated with G(2)-M arrest and the absence of any detectable markers of apoptosis (i.e., plasma membrane blebbing, procaspase 3 activation, loss of mitochondrial membrane potential, and formation of condensed chromatin). Drug-treated cells increased in size and eventually exhibited the characteristics of autophagy (i.e., appearance of autophagosomes and conversion of microtubule-associated protein light chain 3-I to 3-II). The absence of apoptosis was not related to an inhibition of the apoptotic program. Cultures treated with XK469 or SH80 readily underwent apoptosis upon exposure to the Bcl-2/Bcl-x(L) antagonist ethyl 2-amino-6-bromo-4-(1-cyano-2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate. Continued incubation of drug-treated cells led to a reciprocal loss of large autophagic cells and the appearance of smaller cells that could not be stained with Höechst dye HO33342, had a chaotic morphology, were trypan blue-permeable, and lacked mitochondrial membrane potential. L1210 cells cotreated with the phosphatidylinositol-3-kinase inhibitor wortmannin, or having reduced Atg7 protein content, underwent G(2)-M arrest, but not autophagy, following XK469 treatment. Hence, the therapeutic actions of XK469/SH80 with L1210 cultures reflect both the initiation of a cell cycle arrest as well as the initiation of autophagy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Leucemia/patología , Propionatos/farmacología , Quinoxalinas/farmacología , Androstadienos/farmacología , Animales , Proteína 7 Relacionada con la Autofagia , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Vesículas Citoplasmáticas/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Silenciador del Gen , Cinética , Ratones , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Péptido Hidrolasas/metabolismo , Propionatos/química , Quinoxalinas/química , Ensayo de Tumor de Célula Madre , Vacuolas/efectos de los fármacos , Vacuolas/ultraestructura , Wortmanina
17.
J Med Chem ; 50(14): 3274-82, 2007 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-17555307

RESUMEN

Certain farnesyl diphosphate (FPP) analogs are potent inhibitors of the potential anticancer drug target protein farnesyltransferase (FTase), but these compounds are not suitable as drug candidates. Thus, phosphoramidate prodrug derivatives of the monophosphate precursors of FPP-based FTase inhibitors have been synthesized. The monophosphates themselves were significantly more potent inhibitors of FTase than the corresponding FPP analogs. The effects of the prodrug 5b (a derivative of 3-allylfarnesyl monophosphate) have been evaluated on prenylation of RhoB and on the cell cycle in a human malignant schwannoma cell line (STS-26T). In combination treatments, 1-3 microM 5b plus 1 microM lovastatin induced a significant inhibition of RhoB prenylation, and a combination of these drugs at 1 microM each also resulted in significant cell cycle arrest in G1. Indeed, combinations as low as 50 nM lovastatin + 1 microM 5c or 250 nM lovastatin + 50 nM 5c were highly cytostatic in STS-26T cell culture.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/antagonistas & inhibidores , Fosfatos de Poliisoprenilo/síntesis química , Fosfatos de Poliisoprenilo/farmacología , Profármacos/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Espectroscopía de Resonancia Magnética , Profármacos/síntesis química , Espectrometría de Masa por Ionización de Electrospray
18.
Cancer Lett ; 249(2): 294-9, 2007 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-17055152

RESUMEN

L1210 murine leukemia cells exposed to an LD(90) concentration of the Bcl-2/Bcl-x(L) antagonist HA14-1 rapidly undergo apoptosis but also develop numerous intracellular vacuoles with double membranes, exhibit enhanced labeling by monodansylcadaverine, and convert the cytosolic protein LC3-I to LC3-II. These are hallmarks of autophagy. Autophagic vacuoles develop rapidly, preceding the appearance of an apoptotic nuclear morphology and can be observed in both non-apoptotic and apoptotic cells. Inhibition of autophagy by the PI 3-kinase inhibitor wortmannin promoted apoptosis; conversely inhibition of caspase-3/7 with zDEVD-fmk promoted autophagy. Neither process was dependent on calcium translocation. These results indicate that pharmacological suppression of Bcl-2 function can mimic the induction of autophagy that can occur following the down-regulation of Bcl-2 expression by molecular approaches.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Benzopiranos/farmacología , Nitrilos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Microscopía Electrónica
19.
Photochem Photobiol ; 83(5): 1024-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17880495

RESUMEN

Photodynamic therapy (PDT) can cause lethal photodamage by both direct and indirect mechanisms. Direct modes of cell death relate to nonspecific necrosis and the initiation of signaling pathways that elicit apoptosis, autophagy or both. In this report, effects of low-dose and high-dose PDT are explored, comparing sensitizers that localize in the endoplasmic reticulum (the porphycene termed CPO) or mitochondria (mesochlorin). To explore the role of autophagy, two cell lines were examined--the murine L1210 leukemia and an Atg7 knockdown derivative of L1210. The Atg7 gene is central to the process of autophagy. High-dose PDT with either sensitizer resulted in a substantial loss of the Bcl-2 protein. As Bcl-2 regulates both apoptosis and autophagy, loss of this protein can lead to initiation of either or both processes. Low-dose PDT with either sensitizer resulted in the initiation of apoptosis in the L1210/Atg7- cell line and a 20% loss of viability. In contrast, the same PDT dose led to the rapid appearance of autophagic cells in the L1210 line, less apoptosis and only a 5% loss of viability. These results are consistent with autophagy serving as a pro-survival response via the recycling of damaged organelles. At a higher PDT dose more apoptosis was again seen in the L1210/Atg7- line, but both cell lines exhibited comparable cytotoxicity in colony formation assays. We conclude that autophagy offers protection from the phototoxic effects of low-dose PDT, but can serve as an alternate death mode when the PDT dose is increased.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Fármacos Fotosensibilizantes/toxicidad , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Mesoporfirinas/farmacología , Ratones , Fotoquimioterapia , Porfirinas/farmacología
20.
Photochem Photobiol ; 93(6): 1502-1508, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28696570

RESUMEN

We previously reported that a low level of lysosomal photodamage potentiated the phototoxic effect of subsequent mitochondrial photodamage mediated by the benzoporphyrin derivative (BPD) in murine hepatoma 1c1c7 cells. This was attributed to release of Ca2+ from damaged lysosomes and a calpain-mediated conversion of the autophagy-related protein ATG5 to a pro-apoptotic fragment. We now report a comparison of these results with those obtained with the human non-small-cell lung cancer A549 cell line. A549 cells contained lower levels of ATG5 and were less responsive than 1c1c7 cultures to the PDT combination. A rapid appearance of caspase 3/7 activation together with formation of condensed chromatin indicated initiation of apoptosis in both cell lines, but to a lesser extent in A549 cultures. Both cell lines became highly vacuolated within 16 h of combination PDT or an equivalent phototoxic dose from BPD alone. The vacuole periphery was labeled with a fluorescent probe for the endoplasmic reticulum (ER), and vacuole formation was prevented by presence of the protein synthesis inhibitor cycloheximide. These effects are characteristics of a caspase-independent death mode termed paraptosis previously associated with ER stress. These studies suggest that paraptosis may be a more frequent outcome of PDT than has hitherto been realized.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Lisosomas/efectos de la radiación , Mitocondrias/efectos de la radiación , Fotólisis , Apoptosis , Western Blotting , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA