Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Anticancer Drugs ; 33(1): e525-e533, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34387600

RESUMEN

Cancer is related to the cellular proliferative state. Increase in cell-cycle regulatory function augments cellular folate pool. This pathway is therapeutically targeted. A number of drugs influences this metabolism, that is, folic acid, folinic acid, nolatrexed, and methotrexate. Our previous study showed methotrexate influences on rat/human sulfotransferases. Present study explains the effect of nolatrexed (widely used in different cancers) and some micronutrients on the expressions of rat/human sulfotransferases. Female Sprague-Dawley rats were treated with nolatrexed (01-100 mg/kg) and rats of both sexes were treated to folic acid (100, 200, or 400 mg/kg) for 2-weeks and their aryl sulfotransferase-IV (AST-IV; ß-napthol sulfation) and sulfotransferase (STa; DHEA sulfation) activities, protein expression (western blot) and mRNA expression (RT-PCR) were tested. In human-cultured hepatocarcinoma (HepG2) cells nolatrexed (1 nM-1.2 mM) or folinic acid (10 nM-10 µM) were applied for 10 days. Folic acid (0-10 µM) was treated to HepG2 cells. PPST (phenol catalyzing), MPST (dopamine and monoamine), DHEAST (dehydroepiandrosterone and DHEA), and EST (estradiol sulfating) protein expressions (western-blot) were tested in HepG2 cells. Present results suggest that nolatrexed significantly increased sulfotransferases expressions in rat (protein, STa, F = 4.87, P < 0.05/mRNA, AST-IV, F = 6.702, P < 0.014; Student's t test, P < 0.01-0.05) and HepG2 cells. Folic acid increased sulfotransferases activity/protein in gender-dependant manner. Both folic and folinic acid increased several human sulfotransferases isoforms with varied level of significance (least or no increase at highest dose) in HepG2 cells pointing its dose-dependent multiphasic responses. The clinical importance of this study may be furthered in the verification of sulfation metabolism of several exogenous/endogenous molecules, drug-drug interaction and their influences on cancer pathophysiological processes. Further studies are necessary.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Micronutrientes/farmacología , Quinazolinas/farmacología , Sulfotransferasas/efectos de los fármacos , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Arilsulfotransferasa/efectos de los fármacos , Western Blotting , Ciclo Celular , Relación Dosis-Respuesta a Droga , Femenino , Ácido Fólico/administración & dosificación , Ácido Fólico/farmacología , Células Hep G2 , Humanos , Leucovorina/administración & dosificación , Leucovorina/farmacología , Masculino , Metotrexato/administración & dosificación , Metotrexato/farmacología , Micronutrientes/administración & dosificación , Quinazolinas/administración & dosificación , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Factores Sexuales
2.
Artículo en Inglés | MEDLINE | ID: mdl-32315953

RESUMEN

Human schistosomiasis is a disease which globally affects over 229 million people. Three major species affecting humans are Schistosoma mansoni, S. haematobium and S. japonicum. Previous treatment of S. mansoni includes the use of oxamniquine (OXA), a prodrug that is enzymatically activated in S. mansoni but is ineffective against S. haematobium and S. japonicum. The OXA activating enzyme was identified and crystallized, as being a S. mansoni sulfotransferase (SmSULT). S. haematobium and S. japonicum possess homologs of SmSULT (ShSULT and SjSULT) begging the question; why does oxamniquine fail to kill S. haematobium and S. japonicum adult worms? Investigation of the molecular structures of the sulfotransferases indicates that structural differences, specifically in OXA contact residues, do not abrogate OXA binding in the active sites as previously hypothesized. Data presented argue that the ability of SULTs to sulfate and thus activate OXA and its derivatives is linked to the ability of OXA to fit in the binding pocket to allow the transfer of a sulfur group.


Asunto(s)
Oxamniquina/farmacología , Schistosoma/efectos de los fármacos , Sulfotransferasas/química , Animales , Estructura Molecular , Schistosoma/metabolismo , Schistosoma haematobium/efectos de los fármacos , Schistosoma haematobium/metabolismo , Schistosoma japonicum/efectos de los fármacos , Schistosoma japonicum/metabolismo , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/metabolismo , Esquistosomicidas/farmacología , Sulfotransferasas/efectos de los fármacos , Sulfotransferasas/metabolismo
3.
Mol Biochem Parasitol ; 236: 111257, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32027942

RESUMEN

Hycanthone (HYC) is a retired drug formerly used to treat schistosomiasis caused by infection from Schistosoma mansoni and S. haematobium. Resistance to HYC was first observed in S. mansoni laboratory strains and in patients in the 1970s and the use of this drug was subsequently discontinued with the substitution of praziquantel (PZQ) as the single antischistosomal drug in the worldwide formulary. In endemic regions, multiple organizations have partnered with the World Health Organization to deliver PZQ for morbidity control and prevention. While the monotherapy reduces the disease burden, additional drugs are needed to use in combination with PZQ to stay ahead of potential drug resistance. HYC will not be reintroduced into the schistosomiasis drug formulary as a combination drug because it was shown to have adverse properties including mutagenic, teratogenic and carcinogenic activities. Oxamniquine (OXA) was used to treat S. mansoni infection in Brazil during the brief period of HYC use, until the 1990s. Its antischistosomal efficacy has been shown to work through the same mechanism as HYC and it does not possess the undesirable properties linked to HYC. OXA demonstrates cross-resistance in Schistosoma strains with HYC resistance and both are prodrugs requiring metabolic activation in the worm to toxic sulfated forms. The target activating enzyme has been identified as a sulfotransferase enzyme and is currently used as the basis for a structure-guided drug design program. Here, we characterize the sulfotransferases from S. mansoni and S. haematobium in complexes with HYC to compare and contrast with OXA-bound sulfotransferase crystal structures. Although HYC is discontinued for antischistosomal treatment, it can serve as a resource for design of derivative compounds without contraindication.


Asunto(s)
Hicantona , Oxamniquina/análogos & derivados , Esquistosomiasis/tratamiento farmacológico , Sulfotransferasas , Animales , Cristalización/métodos , Cristalografía por Rayos X/métodos , Diseño de Fármacos , Resistencia a Medicamentos , Humanos , Hicantona/efectos adversos , Hicantona/análogos & derivados , Hicantona/química , Oxamniquina/química , Oxamniquina/uso terapéutico , Praziquantel/uso terapéutico , Unión Proteica/efectos de los fármacos , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Schistosoma haematobium/efectos de los fármacos , Schistosoma haematobium/metabolismo , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/metabolismo , Esquistosomicidas/uso terapéutico , Sulfotransferasas/efectos de los fármacos , Sulfotransferasas/metabolismo
4.
Mol Biol Rep ; 45(6): 2571-2584, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30315444

RESUMEN

N-ethyl-N-nitrosourea (ENU) is highly used in rodent models of tumerogenesis/carcinogenesis. Xenografting human-cancer tissues/cells with estradiol (E2) treatment is also used to generate rodent-models of gynaecological cancers. The altered metabolic-redox environment leading to establishment of pre-tumorigenesis condition and their mechanism are less studied. Here, female Wister rats were treated with these drugs at their pre-tumerogenic dosage (one group ENU single intra-peritoneal dose of 90 mg/kg b.w. and another group were implanted with human breast tumor (stage-IIIB) and fed with 2.5 mg of 17ß-estradiol once in a week for 4 months). After 4 months, animals were sacrificed; their serum and liver tissues were tested. A brief comparison was made with a rat model (regarded as positive control) of toxicity induced by mutagenic environmental pollutant arsenic (0.6 ppm daily/4 weeks). The increase in serum alkaline phosphatase and glutamate-pyruvate transaminase suggests the possible organ toxicity is favoured by the increase in hepatic/systemic free radicals and oxidative stress in all drug application models. But the increase in the serum E2 level as noted in the ELISA data with impairment in the hepatic estrogen sulfotransferase (SULT1E1) protein expression (immuno-blot data) were noticed with interfered hepatic free-thiols only in ENU and xenograft-E2 group compared to arsenic group. It is also evident in the in vitro result from E2/GSH/NAC added hepatic slices with altered antioxidant regulations. Moreover, impairment in hepatic SOD1, catalase and glutathiole peroxidase activities (PAGEzymographic data), especially in the ENU-treated group makes them more vulnerable to the oxidative threat in creating pre-tumerogenic microenvironment. This is evident in the result of their higher DNA-damage and histological abnormalities. The Bioinformatics study revealed an important role of rSULT1E1 in the regulations of E2 metabolism. This study is important for the exploration of the pre-tumerogenic condition by ENU and E2 by impairing SULT1E1 expression and E2 regulations via oxidant-stress signalling. The finding may help to find new therapeutic-targets to treat gynaecological-cancers more effectively.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Estradiol/farmacología , Etilnitrosourea/farmacología , Animales , Antioxidantes/metabolismo , Neoplasias de la Mama/metabolismo , Catalasa/efectos de los fármacos , Catalasa/metabolismo , Daño del ADN/efectos de los fármacos , Estradiol/sangre , Estradiol/metabolismo , Etilnitrosourea/metabolismo , Femenino , Xenoinjertos , Humanos , Hígado/metabolismo , Oxidantes/metabolismo , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Sulfotransferasas/efectos de los fármacos , Sulfotransferasas/genética , Superóxido Dismutasa-1/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Transl Psychiatry ; 7(3): e1061, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28291260

RESUMEN

The neurotrophic hypothesis of depression suggests an association between effects on neuroplasticity and clinical response to antidepressant drug therapy. We studied individual variability in antidepressant drug effects on cell proliferation in lymphoblastoid cell lines (LCLs) from n=25 therapy-resistant patients versus n=25 first-line therapy responders from the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study. Furthermore, the variability in gene expression of genes associated with cell proliferation was analyzed for tentative candidate genes for prediction of individual LCL donor's treatment response. Cell proliferation was quantified by EdU (5-ethynyl-2'-deoxyuridine) assays after 21-day incubation of LCLs with fluoxetine (0.5 ng µl-1) and citalopram (0.3 ng µl-1) as developed and described earlier. Gene expression of a panel of candidate genes derived from genome-wide expression analyses of antidepressant effects on cell proliferation of LCLs from the Munich Antidepressant Response Signature (MARS) study was analyzed by real-time PCR. Significant differences in in vitro cell proliferation effects were detected between the group of LCLs from first-line therapy responders and LCLs from treatment-resistant patients. Gene expression analysis of the candidate gene panel revealed and confirmed influence of the candidate genes ABCB1, FZD7 and WNT2B on antidepressant drug resistance. The potential of these genes as tentative biomarkers for antidepressant drug resistance was confirmed. In vitro cell proliferation testing may serve as functional biomarker for individual neuroplasticity effects of antidepressants.


Asunto(s)
Antidepresivos/farmacología , Proliferación Celular/efectos de los fármacos , Trastorno Depresivo Resistente al Tratamiento/genética , Células Progenitoras Linfoides/efectos de los fármacos , Subfamilia B de Transportador de Casetes de Unión a ATP/efectos de los fármacos , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Adulto , Antidepresivos/uso terapéutico , Biomarcadores , Línea Celular , Proliferación Celular/genética , Citalopram/farmacología , Citalopram/uso terapéutico , Trastorno Depresivo Resistente al Tratamiento/tratamiento farmacológico , Femenino , Fluoxetina/farmacología , Receptores Frizzled/efectos de los fármacos , Receptores Frizzled/genética , Glicoproteínas/efectos de los fármacos , Glicoproteínas/genética , Humanos , Técnicas In Vitro , Células Progenitoras Linfoides/metabolismo , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Sulfotransferasas/efectos de los fármacos , Sulfotransferasas/genética , Proteína 2 Similar al Factor de Transcripción 7/efectos de los fármacos , Proteína 2 Similar al Factor de Transcripción 7/genética , Transcriptoma , Proteínas Wnt/efectos de los fármacos , Proteínas Wnt/genética
6.
J Pharmacol Exp Ther ; 355(3): 429-41, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26427720

RESUMEN

Cytosolic sulfotransferase 1C2 (SULT1C2) is expressed in the kidney, stomach, and liver of rats; however, the mechanisms regulating expression of this enzyme are not known. We evaluated transcriptional regulation of SULT1C2 by mevalonate (MVA)-derived intermediates in primary cultured rat hepatocytes using several cholesterol synthesis inhibitors. Blocking production of mevalonate with the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor pravastatin (30 µM), reduced SULT1C2 mRNA content by ∼40% whereas the squalene synthase inhibitor squalestatin (SQ1, 0.1 µM), which causes accumulation of nonsterol isoprenoids, increased mRNA content by 4-fold. Treatment with MVA (10 mM) strongly induced SULT1C2 mRNA by 12-fold, and this effect was blocked by inhibiting squalene epoxidase but not by more distal cholesterol inhibitors, indicating the effects of MVA are mediated by postsqualene metabolites. Using rapid amplification of cDNA ends (RACE), we characterized the 5' end of SULT1C2 mRNA and used this information to generate constructs for promoter analysis. SQ1 and MVA increased reporter activity by ∼1.6- and 3-fold, respectively, from a construct beginning 49 base pairs (bp) upstream from the longest 5'-RACE product (-3140:-49). Sequence deletions from this construct revealed a hepatocyte nuclear factor 1 (HNF1) element (-2558), and mutation of this element reduced basal (75%) and MVA-induced (30%) reporter activity and attenuated promoter activation following overexpression of HNF1α or 1ß. However, the effects of SQ1 were localized to a more proximal promoter region (-281:-49). Collectively, our findings demonstrate that cholesterol biosynthetic intermediates influence SULT1C2 expression in rat primary hepatocytes. Further, HNF1 appears to play an important role in mediating basal and MVA-induced SULT1C2 transcription.


Asunto(s)
Colesterol/biosíntesis , Regulación Enzimológica de la Expresión Génica/fisiología , Hepatocitos/enzimología , Sulfotransferasas/biosíntesis , Sulfotransferasas/genética , Animales , Anticolesterolemiantes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Factor Nuclear 1 del Hepatocito/genética , Factor Nuclear 1 del Hepatocito/metabolismo , Hepatocitos/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Masculino , Ácido Mevalónico/farmacología , Cultivo Primario de Células , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Escualeno-Monooxigenasa/antagonistas & inhibidores , Sulfotransferasas/efectos de los fármacos , Transfección , Ácidos Tricarboxílicos/farmacología
7.
Gynecol Endocrinol ; 31(7): 578-81, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26036805

RESUMEN

Critical steps of embryo implantation are controlled by progesterone. These processes can be interrupted by progesterone receptor (PR) antagonists, e.g. drugs used for abortion. Antiprogestin effects induced by natural compounds and environmental chemicals have been rarely addressed. In our in vitro study, we investigated putative antiprogestin activities of the plant compounds apigenin (API) and trans-ferulic acid (t-FA) as well as the UV absorbers octyl methoxycinnamate (OMC) and 4-methylbenzylidene camphor (4-MBC). They were compared with the selective progesterone receptor modulators (SPRMs) mifepristone (RU486) and ulipristal acetate (UPA) as well as the full PR-antagonist ZK137316. Effects of test compounds in combination with progesterone on the progesterone-sensitive target gene estrogen sulfotransferase (SULT1E1) were characterized by sigmoidal concentration-response curves obtained by RT-qPCR. The agonistic effect of progesterone on SULT1E1 mRNA levels was concentration-dependently antagonized by RU486, UPA and ZK137316 as well as, with lower potency, apigenin. t-FA, OMC and 4-MBC had no effect on SULT1E1 mRNA levels. We demonstrated that apigenin, although at higher concentrations, exerts a similar effect as the well-characterized SPRMs RU486 and UPA or the progesterone antagonist ZK137316 in this model. Our endometrium-specific Ishikawa cell assay is a useful complement to artificial transactivation assays for the identification of environmental substances with antiprogestin activities.


Asunto(s)
Endometrio , Antagonistas de Hormonas/farmacología , Fitoquímicos/farmacología , Receptores de Progesterona/antagonistas & inhibidores , Sulfotransferasas/efectos de los fármacos , Protectores Solares/farmacología , Apigenina/farmacología , Alcanfor/análogos & derivados , Alcanfor/farmacología , Línea Celular Tumoral , Ácidos Cumáricos/farmacología , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Femenino , Humanos , Mifepristona/farmacología , Norpregnadienos/farmacología
8.
Mol Cell Biochem ; 357(1-2): 83-94, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21594647

RESUMEN

Malignant pleural mesothelioma (MPM) is an aggressive, asbestos-related malignancy of the thoracic pleura. Although, platinum-based agents are the first line of therapy, there is an urgent need for second-line therapies to treat the drug-resistant MPM. Cell cycle as well as apoptosis pathways are frequently altered in MPM and thus remain attractive targets for intervention strategies. Curcumin, the major component in the spice turmeric, alone or in combination with other chemotherapeutics has been under investigation for a number of cancers. In this study, we investigated the biological and molecular responses of MPM cells to curcumin treatments and the mechanisms involved. Flow-cytometric analyses coupled with western immunoblotting and gene-array analyses were conducted to determine mechanisms of curcumin-dependent growth suppression of human (H2373, H2452, H2461, and H226) and murine (AB12) MPM cells. Curcumin inhibited MPM cell growth in a dose- and time-dependent manner while pretreatment of MPM cells with curcumin enhanced cisplatin efficacy. Curcumin activated the stress-activated p38 kinase, caspases 9 and 3, caused elevated levels of proapoptotic proteins Bax, stimulated PARP cleavage, and apoptosis. In addition, curcumin treatments stimulated expression of novel transducers of cell growth suppression such as CARP-1, XAF1, and SULF1 proteins. Oral administration of curcumin inhibited growth of murine MPM cell-derived tumors in vivo in part by stimulating apoptosis. Thus, curcumin targets cell cycle and promotes apoptosis to suppress MPM growth in vitro and in vivo. Our studies provide a proof-of-principle rationale for further in-depth analysis of MPM growth suppression mechanisms and their future exploitation in effective management of resistant MPM.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Curcumina/farmacología , Mesotelioma/metabolismo , Neoplasias Pleurales/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/efectos de los fármacos , Caspasas/efectos de los fármacos , Proteínas de Ciclo Celular , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Ratones , Proteínas de Neoplasias/efectos de los fármacos , Sulfotransferasas/efectos de los fármacos , Proteína X Asociada a bcl-2/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
9.
FASEB J ; 24(2): 436-50, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19812376

RESUMEN

Heparan sulfate proteoglycans (HSPGs), strategically located at the cell-tissue-organ interface, regulate major biological processes, including cell proliferation, migration, and adhesion. These vital functions are compromised in tumors, due, in part, to alterations in heparan sulfate (HS) expression and structure. How these modifications occur is largely unknown. Here, we investigated whether epigenetic abnormalities involving aberrant DNA methylation affect HS biosynthetic enzymes in cancer cells. Analysis of the methylation status of glycosyltransferase and sulfotransferase genes in H-HEMC-SS chondrosarcoma cells showed a typical hypermethylation profile of 3-OST sulfotransferase genes. Exposure of chondrosarcoma cells to 5-aza-2'-deoxycytidine (5-Aza-dc), a DNA-methyltransferase inhibitor, up-regulated expression of 3-OST1, 3-OST2, and 3-OST3A mRNAs, indicating that aberrant methylation affects transcription of these genes. Furthermore, HS expression was restored on 5-Aza-dc treatment or reintroduction of 3-OST expression, as shown by indirect immunofluorescence microscopy and/or analysis of HS chains by anion-exchange and gel-filtration chromatography. Notably, 5-Aza-dc treatment of HEMC cells or expression of 3-OST3A cDNA reduced their proliferative and invading properties and augmented adhesion of chondrosarcoma cells. These results provide the first evidence for specific epigenetic regulation of 3-OST genes resulting in altered HSPG sulfation and point to a defect of HS-3-O-sulfation as a factor in cancer progression.


Asunto(s)
Metilación de ADN , Proteoglicanos de Heparán Sulfato/biosíntesis , Sulfotransferasas/genética , Azacitidina/análogos & derivados , Azacitidina/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Condrosarcoma/genética , Islas de CpG/fisiología , Metilación de ADN/efectos de los fármacos , Decitabina , Represión Enzimática , Células HL-60 , Células HeLa , Proteoglicanos de Heparán Sulfato/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Invasividad Neoplásica/genética , Sulfotransferasas/efectos de los fármacos , Regulación hacia Arriba
10.
Biol Pharm Bull ; 32(1): 105-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19122289

RESUMEN

Sulfotransferase 1A3 (SULT1A3) is a phase II detoxifying enzyme of xenobiotics predominantly expressed in the intestinal epithelium. Recent increase in the use of herbal extracts as dietary supplements may lead to an increase in the possibility of dietary supplement-drug interactions. The purpose of the present study was to investigate the effects of 18 herbal extracts on SULT1A3 activity and the possibility of interaction between medicinal drugs and herbal extracts. We examined the inhibitory potencies of 18 herbal extracts on the sulfation of dopamine, a typical substrate of SULT1A3, and ritodrine, a beta(2) stimulant, by human recombinant SULT1A3. The sulfation of dopamine was inhibited by extracts of banaba, green tea, Rafuma, grape seed, peanut seed coat, gingko biloba leaf, St. John's wort, gymnema and milkthistle. The IC(50) values of these herbal extracts were lower than the putative gastrointestinal concentration when the recommended dose was ingested. On the other hand, chlorella extract and rutin showed no inhibitory effects and wheat, mulberry and siberian ginseng had IC(50) values exceedingly higher than the putative gastrointestinal concentration. The inhibitory profiles of herbal extracts for the sulfation of ritodrine were comparable to those for the sulfation of dopamine. In conclusion, the extracts of herbs such as banaba and green tea potently inhibited SULT1A3 activity. These extracts may increase the bioavailability of drugs whose bioavailabilities were limited by the function of SULT1A3 on the intestinal epithelium.


Asunto(s)
Interacciones de Hierba-Droga , Extractos Vegetales/farmacología , Sulfotransferasas/efectos de los fármacos , Análisis de Varianza , Arilsulfotransferasa , Humanos , Concentración 50 Inhibidora , Proteínas Recombinantes/farmacología
11.
J Med Food ; 11(4): 623-8, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19053852

RESUMEN

Several fruit juices have been reported to cause food-drug interactions, mainly affecting cytochrome P450 activity; however, little is known about the effects of fruit juices on conjugation reactions. Among several fruit juices tested (apple, peach, orange, pineapple, grapefruit, and pomegranate), pomegranate juice potently inhibited the sulfoconjugation of 1-naphthol in Caco-2 cells. This inhibition was both dose- and culture time-dependent, with a 50% inhibitory concentration (IC(50)) value calculated at 2.7% (vol/vol). In contrast, no obvious inhibition of glucuronidation of 1-naphthol in Caco-2 cells was observed by any of the juices examined. Punicalagin, the most abundant antioxidant polyphenol in pomegranate juice, was also found to strongly inhibit sulfoconjugation in Caco-2 cells with an IC(50) of 45 microM, which is consistent with that of pomegranate juice. These data suggest that punicalagin is mainly responsible for the inhibition of sulfoconjugation by pomegranate juice. We additionally demonstrated that pomegranate juice and punicalagin both inhibit phenol sulfotransferase activity in Caco-2 cells in vitro, at concentrations that are almost equivalent to those used in the Caco-2 cells. Pomegranate juice, however, shows no effects on the expression of the sulfotransferase SULT1A family of genes (SULT1A1 and SULT1A3) in Caco-2 cells. These results indicate that the inhibition of sulfotransferase activity by punicalagin in Caco-2 cells is responsible for the reductions seen in 1-naphthyl sulfate accumulation. Our data also suggest that constituents of pomegranate juice, most probably punicalagin, impair the enteric functions of sulfoconjugation and that this might have effects upon the bioavailability of drugs and other compounds present in food and in the environment. These effects might be related to the anticarcinogenic properties of pomegranate juice.


Asunto(s)
Neoplasias del Colon/metabolismo , Interacciones Alimento-Droga/fisiología , Taninos Hidrolizables/farmacología , Lythraceae , Preparaciones de Plantas , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/efectos de los fármacos , Antioxidantes/farmacología , Bebidas , Células CACO-2 , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Taninos Hidrolizables/aislamiento & purificación , Mucosa Intestinal/efectos de los fármacos , Fase I de la Desintoxicación Metabólica , Naftoles/farmacocinética , Proteínas de Neoplasias/efectos de los fármacos , Sulfotransferasas/efectos de los fármacos
12.
Drug Metab Lett ; 2(2): 115-119, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19356074

RESUMEN

Our earlier investigation showed that MTX is an inducer of rat and human sulfotransferases. Here we report that folic acid treatment inhibited MTX induction of aryl sulfotransferase (AST-IV) in female rat liver and hydroxysteroid sulfotransferase (STa) in male rat liver. This is important for understanding the clinical mechanisms of MTX.


Asunto(s)
Arilsulfotransferasa/efectos de los fármacos , Ácido Fólico/efectos de los fármacos , Metotrexato/farmacología , Sulfotransferasas/efectos de los fármacos , Animales , Antimetabolitos Antineoplásicos/farmacología , Arilsulfotransferasa/metabolismo , Interacciones Farmacológicas , Inducción Enzimática/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/enzimología , Hígado/metabolismo , Masculino , ARN Mensajero/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factores Sexuales , Sulfotransferasas/metabolismo
13.
Drug Metab Lett ; 2(3): 198-204, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19356094

RESUMEN

We used protein-ligand docking and minimization to identify celecoxib as an allosteric modulator of SULT2A1-catalyzed estradiol sulfonation. Subsequent to celecoxib docking and complex minimization, conformational changes in SULT2A1 allowed estradiol docking to an alternative binding region with predicted preference for 17beta-OH-E(2) sulfonation over 3-OH-E(2) sulfonation.


Asunto(s)
Estradiol/metabolismo , Pirazoles/farmacología , Sulfonamidas/farmacología , Sulfotransferasas/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Sitios de Unión/efectos de los fármacos , Celecoxib , Interacciones Farmacológicas , Humanos , Ligandos , Unión Proteica , Conformación Proteica/efectos de los fármacos , Programas Informáticos , Sulfotransferasas/genética
14.
Life Sci ; 81(25-26): 1659-67, 2007 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-17963788

RESUMEN

Sulfotransferases (SULTs) play an important role in the detoxification and bioactivation of endogenous compounds and xenobiotics. Studies on rat sulfotransferases had shown that SULT genes, like cytochrome P450 genes, can be regulated by ligands that bind nuclear receptors. For human SULT genes, the regulation of human SULT2A1 expression is currently the best characterized. In this study, we systematically examined the regulation of human SULT1A genes by glucocorticoids. Treatment of the human hepatocellular carcinoma derived HepG2 cells with 10(-7) M dexamethasone did not affect the SULT1A1 activity toward p-nitrophenol. In contrast, SULT1A3 activity toward dopamine was significantly induced. Transient transfection of the SULT1A3 5'-flanking region/luciferase reporter construct showed that SULT1A3 was responsive to dexamethasone and prednisolone in a concentration-dependent manner with maximal induction at 10(-7) M dexamethasone or 1 microM prednisolone. In addition, induction by dexamethasone was dependent on the level of expression of the glucocorticoid receptor. Analysis of the 5'-flanking region led to the identification of a putative glucocorticoid response element at position (-1211 to -1193) upstream of the transcription start site and deletion or mutation of this element resulted in a loss of response. In summary, the data from this study shows that the human SULT1A3 gene is inducible by glucocorticoids through a glucocorticoid receptor-mediated mechanism and the glucocorticoid response element at position (-1211 to -1193) is necessary for this induction.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Sulfotransferasas/genética , Animales , Dexametasona/metabolismo , Dexametasona/farmacología , Hepatocitos/metabolismo , Humanos , Luciferasas , Prednisolona/metabolismo , Prednisolona/farmacología , Ratas , Receptores de Glucocorticoides/genética , Elementos de Respuesta/genética , Sulfotransferasas/efectos de los fármacos , Transfección , Células Tumorales Cultivadas
15.
Biopharm Drug Dispos ; 28(9): 491-500, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17876860

RESUMEN

Sulfotransferase (SULT) 1A1 and SULT1A3 play important roles in the presystemic inactivation of beta(2) agonists in the liver and intestine, respectively. The study aimed to investigate the inhibitory effects of grapefruit juice, orange juice, green tea, black tea and oolong tea and their constituents on the activities of SULT1A1 and SULT1A3. The activities of both SULT1A1 and SULT1A3 were significantly inhibited by all the beverages investigated at a concentration of 10%. The beverage constituents were tested in concentration ranges considered to be physiologically relevant. The grapefruit constituent, quercetin, completely inhibited SULT1A1, while quercetin and naringin both partially inhibited SULT1A3. The orange constituents, tangeretin and nobiletin, also completely inhibited SULT1A1. The tea constituents, (-)-epicatechin gallate and (-)-epigallocatechin gallate, both almost completely inhibited SULT1A1 and SULT1A3. Moreover, the theaflavin and thearubigin fractions of black tea both completely inhibited SULT1A1 and strongly inhibited SULT1A3. The inhibitory action of green tea on SULT1A3 was competitive, while that of black tea and oolong tea was mixed competitive/non-competitive. Mechanism-based inhibition was not observed with any beverage. In conclusion, various beverages, especially teas, inhibit the function of SULT1A3, and therefore may have the potential to increase the bioavailability of orally administered substrates of SULT1A3, such as beta(2) agonists.


Asunto(s)
Agonistas Adrenérgicos beta/metabolismo , Arilsulfotransferasa/efectos de los fármacos , Bebidas , Sulfotransferasas/efectos de los fármacos , Arilsulfotransferasa/metabolismo , Biflavonoides/farmacología , Disponibilidad Biológica , Catequina/análogos & derivados , Catequina/farmacología , Citrus paradisi/química , Citrus sinensis/química , Flavanonas/farmacología , Flavonas/farmacología , Interacciones de Hierba-Droga , Humanos , Técnicas In Vitro , Fenoles/farmacología , Polifenoles , Quercetina/farmacología , Sulfotransferasas/metabolismo , Té/química
16.
Aquat Toxicol ; 81(3): 286-92, 2007 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-17239972

RESUMEN

The sulfonation of 17beta-estradiol (E2) by human liver and recombinant sulfotransferases is influenced by environmental contaminants such as hydroxylated metabolites of polychlorinated biphenyls (OH-PCBs), which are potent inhibitors, and the therapeutic drug, celecoxib, which affects positional sulfonation of E2. In some locations, the aquatic environment is contaminated by PCBs, OH-PCBs and widely used therapeutic drugs. The objectives of this study were to investigate the sulfonation kinetics of E2 in liver cytosol from channel catfish (Ictalurus punctatus); to examine the effect of OH-PCBs on E2 sulfonation; and to determine if celecoxib altered the position of E2 sulfonation, as it does with human liver cytosol. E2 was converted to both 3- and 17-sulfates by catfish liver cytosol. At E2 concentrations below 1 microM, formation of E2-3-sulfate (E2-3-S) predominated, but substrate inhibition was observed at higher concentrations. Rates of E2-3-S formation at different E2 concentrations were fit to a substrate inhibition model, with K'm and V'max values of 0.40 +/- 0.10 microM and 91.0 +/- 4.7 pmol/min/mg protein, respectively and K(i) of 1.08 +/- 0.09 microM. The formation of E2-17-S fit Michaelis-Menten kinetics over the concentration range 25 nM to 2.5 microM, with K(m) and V(max) values of 1.07 +/- 0.23 microM and 25.7 +/- 4.43 pmol/min/mg protein, respectively. The efficiency (V(max)/K(m)) of formation of E2-3-S was 9.8-fold higher than that of E2-17-S. Several OH-PCBs inhibited E2 3-sulfonation, measured at an E2 concentration of 1 nM. Of those tested, the most potent inhibitor was 4'-OH-CB79, with two chlorine atoms flanking the OH group (IC(50): 94 nM). The inhibition of estrogen sulfonation by OH-PCBs may disrupt the endocrine system and thus contribute to the known toxic effects of these compounds. Celecoxib did not stimulate E2-17-S formation, as is the case with human liver cytosol, but did inhibit the formation of E2-3-S (IC(50): 44 microM) and to a lesser extent, E2-17-S (IC(50): > 160 microM), suggesting the previously found effect of celecoxib on E2-17-S formation may be specific to human SULT2A1.


Asunto(s)
Estradiol/metabolismo , Ictaluridae/metabolismo , Bifenilos Policlorados/toxicidad , Pirazoles/toxicidad , Sulfonamidas/toxicidad , Sulfotransferasas/efectos de los fármacos , Animales , Celecoxib , Citosol/química , Inhibidores Enzimáticos/toxicidad , Estradiol/análisis , Femenino , Concentración 50 Inhibidora , Cinética , Hígado/efectos de los fármacos , Masculino , Bifenilos Policlorados/química , Pirazoles/química , Sulfonamidas/química , Sulfotransferasas/antagonistas & inhibidores
17.
Environ Health Perspect ; 115 Suppl 1: 51-4, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18174950

RESUMEN

BACKGROUND: Sulfation plays an important role both in detoxification and in the control of steroid activity. Studies in rodents have shown that the conversion of dehydroepiandrosterone (DHEA) to DHEA-sulfate is involved in learning and the memory process. METHODS: The effects of a range of plasticizers and related compounds commonly encountered in the environment were evaluated kinetically against human DHEA sulfotransferase (SULT 2A1) and by reverse transcriptase-polymerase chain reaction (RT-PCR) against several enzymes involved in the synthesis of the sulfotransferase cofactor adenosine 3'-phosphate 5'-phosphosulfate (PAPS). RESULTS: We found that several of the chemicals acted as competitive inhibitors of SULT 2A1 (K(i) for 4-tert-octylphenol is 2.8 microM). Additionally, after treatment of TE 671 cells with 0.005-0.5 microM 4-n-octylphenol, bis(2-ethylhexyl)phthalate, and diisodecyl phthalate, real-time RT-PCR showed dose-dependent decreases in the steady-state mRNA levels of cysteine dioxygenase type I, sulfite oxidase, and 3'-phosphate 5'-phosphosulfate synthase I. CONCLUSIONS: These data suggest that environmental contaminants may exert effects on neuronal function both by direct inhibition of sulfotransferase enzymes and by interrupting the supply of PAPS, which has wider implications for endocrine disruption and xenobiotic metabolism.


Asunto(s)
Disruptores Endocrinos/toxicidad , Contaminantes Ambientales/toxicidad , Fosfoadenosina Fosfosulfato/metabolismo , Sulfotransferasas/efectos de los fármacos , Línea Celular Tumoral , Cisteína-Dioxigenasa/efectos de los fármacos , Cisteína-Dioxigenasa/metabolismo , Dietilhexil Ftalato/toxicidad , Relación Dosis-Respuesta a Droga , Humanos , Meduloblastoma/metabolismo , Complejos Multienzimáticos/efectos de los fármacos , Complejos Multienzimáticos/metabolismo , Fenoles/toxicidad , Ácidos Ftálicos/toxicidad , Plastificantes/toxicidad , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfato Adenililtransferasa/efectos de los fármacos , Sulfato Adenililtransferasa/metabolismo , Sulfito-Oxidasa/efectos de los fármacos , Sulfito-Oxidasa/metabolismo , Sulfotransferasas/metabolismo
18.
Chem Biol ; 13(8): 891-7, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16931338

RESUMEN

Bisphenol A (BPA) is a widely used plasticizer whose estrogenic properties may impact hormone-responsive disorders and fetal development. In vivo, BPA appears to have greater activity than is suggested by its estrogen receptor (ER) binding affinity. This may be a result of BPA sulfation/desulfation providing a pathway for selective uptake into hormone-responsive cells. BPA is a substrate for estrogen sulfotransferase, and bisphenol A sulfate (BPAS) and disulfate are substrates for estrone sulfatase. Although the sulfated xenobiotics bind poorly to the ER, both stimulated the growth of receptor-positive breast tumor cells. Treatment of MCF-7 cells with BPAS leads to desulfation and uptake of BPA. No BPAS is found inside the cells. These findings suggest a mechanism for the selective uptake of BPA into cells expressing estrone sulfatase. Therefore, sulfation may increase the estrogenic potential of xenobiotics.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fenoles/farmacocinética , Sulfotransferasas/efectos de los fármacos , Óxidos de Azufre/química , Ésteres del Ácido Sulfúrico/farmacocinética , Compuestos de Bencidrilo , Sitios de Unión , Catálisis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Espectroscopía de Resonancia Magnética/métodos , Estructura Molecular , Fenoles/síntesis química , Fenoles/química , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Sensibilidad y Especificidad , Sulfotransferasas/química , Sulfotransferasas/metabolismo , Ésteres del Ácido Sulfúrico/síntesis química , Ésteres del Ácido Sulfúrico/química
20.
Aquat Toxicol ; 78 Suppl 1: S65-72, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16600398

RESUMEN

Significant amounts of oil and alkylphenols are released into the sea by petroleum installations as a result of discharges of produced water. Some of these pollutants elicit estrogenic responses in fish, but their effects on the endocrine system of molluscs are largely unknown. In this study, mussels Mytilus edulis were exposed to North Sea oil (O) and the mixture of North Sea oil+alkylphenols (OAP), and the effects on tissue steroid levels and steroid metabolism (P450-aromatase and estradiol-sulfotransferase) were monitored. Levels of free testosterone and free estradiol were much higher in gonad tissue than in peripheral tissue, whereas esterified steroids (released after saponification) were of the same order of magnitude in both tissues. Levels of free steroids determined in gonads were not affected by exposure, but esterified steroids significantly increased in OAP exposed mussels (up to 2.4-fold). The sulfation of estradiol was investigated as a conjugation pathway, and increased activities were observed in digestive gland cytosol of both O and OAP exposure groups (up to 2.8-fold). Additionally, increased P450-aromatase activity was determined in OAP exposed mussels (up to three-fold, both in gonad and digestive gland), but not in the O group. Altogether, the results indicate that North Sea oil leads to increased sulfation of estradiol, and that in combination with alkylphenols, additional alterations are observed: increased P450-aromatase, and increased levels of esterified-steroids in gonads. Nonetheless, mussels are able to maintain gonad concentrations of free steroids unaltered, possibly via homeostatic mechanisms such as the conjugation with fatty acid or the formation of sulphate conjugates.


Asunto(s)
Mytilus edulis/efectos de los fármacos , Petróleo/toxicidad , Fenoles/toxicidad , Sulfotransferasas/efectos de los fármacos , Contaminantes Químicos del Agua/toxicidad , Análisis de Varianza , Animales , Aromatasa/metabolismo , Citosol/metabolismo , Sistema Digestivo/efectos de los fármacos , Estradiol/análisis , Estradiol/metabolismo , Gónadas/efectos de los fármacos , Sulfotransferasas/metabolismo , Testosterona/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...